1
|
Lwin HY, Tiskratok W, Kyawsoewin M, Manokawinchoke J, Termkwanchareon C, Limjeerajarus N, Limjeerajarus CN, Egusa H, Osathanon T, Limraksasin P. Shear Stress Regulates Osteogenic Differentiation of Human Dental Pulp Stem Cells via the p38 Pathway. Int J Mol Sci 2025; 26:5667. [PMID: 40565131 PMCID: PMC12193168 DOI: 10.3390/ijms26125667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2025] [Revised: 06/07/2025] [Accepted: 06/11/2025] [Indexed: 06/28/2025] Open
Abstract
This study aimed to investigate the effects of shear stress on osteogenic differentiation of human dental pulp stem cells (hDPSCs). The hDPSCs were subjected to shear stress for 24 h before osteogenic induction for 21 days. The mRNA expression of osteogenic markers such as RUNX2, OSX, ALP, COL1A1, OCN, and OPN was evaluated by real-time RT-PCR. Alkaline Phosphatase (ALP) activity and Alizarin Red S (ARS) staining were investigated to confirm osteogenic differentiation and mineralization of hDPSCs, respectively. The protein expression of osterix was shown by immunofluorescence staining and Western blotting. RNA sequencing was performed to investigate how shear stress affects the osteogenic differentiation of hDPSCs, which was validated through p38 inhibitor (SB203580) treatment. Real-time RT-PCR revealed that shear stress enhanced osteogenic marker-gene expression. The increased osterix protein expression was detected on Day 14 in the shear-stress loading group compared to the static group. Shear stress enhanced ALP activity and mineralization, observed on Days 14 and 21. A volcano plot exhibited up- and downregulated genes, while the p38 inhibitor markedly inhibited osteogenic differentiation of hDPSCs triggered by shear stress. In conclusion, shear stress promotes the osteogenic differentiation of hDPSCs through the p38 mitogen-activated protein kinase signaling pathway.
Collapse
Grants
- HEAF67320037, P.L This study was supported by the Thailand Science Research and Innovation Fund, Chulalongkorn University, the Fundamental Fund 2567
- (H.Y.L.) This research project was supported by the Second Century Fund (C2F), Chulalongkorn University
- (P.L) grants for the development of new faculty staff, Ratchadaphiseksomphot Fund, Chulalongkorn University
- NRIIS195680, W.T. This work was supported by the Suranaree University of Technology (SUT), Thailand Science Research and Innovation (TSRI), and the National Science, Research and Innovation Fund
Collapse
Affiliation(s)
- Hnin Yu Lwin
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (H.Y.L.); (M.K.); (J.M.); (C.T.); (T.O.)
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watcharaphol Tiskratok
- School of Geriatric Oral Health, Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Maythwe Kyawsoewin
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (H.Y.L.); (M.K.); (J.M.); (C.T.); (T.O.)
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jeeranan Manokawinchoke
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (H.Y.L.); (M.K.); (J.M.); (C.T.); (T.O.)
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chutimon Termkwanchareon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (H.Y.L.); (M.K.); (J.M.); (C.T.); (T.O.)
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nuttapol Limjeerajarus
- Office of Academic Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand;
| | | | - Hiroshi Egusa
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (H.Y.L.); (M.K.); (J.M.); (C.T.); (T.O.)
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Miyagi, Japan
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Miyagi, Japan
| | - Thanaphum Osathanon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (H.Y.L.); (M.K.); (J.M.); (C.T.); (T.O.)
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Phoonsuk Limraksasin
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (H.Y.L.); (M.K.); (J.M.); (C.T.); (T.O.)
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Miyagi, Japan
| |
Collapse
|
2
|
Aung HH, Pothipan P, Aswakool J, Santironnarong S, Phatthanakun R, Pinrod V, Jiemsakul T, Chancharoen W, Moonwiriyakit A. Non-invasive measurement of wall shear stress in microfluidic chip for osteoblast cell culture using improved depth estimation of defocus particle tracking method. BIOMICROFLUIDICS 2024; 18:054114. [PMID: 39464242 PMCID: PMC11510738 DOI: 10.1063/5.0226294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
The development of a non-invasive method for measuring the internal fluid behavior and dynamics of microchannels in microfluidics poses critical challenges to biological research, such as understanding the impact of wall shear stress (WSS) in the growth of a bone-forming osteoblast. This study used the General Defocus Particle Tracking (GDPT) technique to develop a non-invasive method for quantifying the fluid velocity profile and calculated the WSS within a microfluidic chip. The GDPT estimates particle motion in a three-dimensional space by analyzing two-dimensional images and video captured using a single camera. However, without a lens to introduce aberration, GDPT is prone to error in estimating the displacement direction for out-of-focus particles, and without knowing the exact refractive indices, the scaling from estimated values to physical units is inaccurate. The proposed approach addresses both challenges by using theoretical knowledge on laminar flow and integrating results obtained from multiple analyses. The proposed approach was validated using computational fluid dynamics (CFD) simulations and experimental video of a microfluidic chip that can generate different WSS levels under steady-state flow conditions. By comparing the CFD and GDPT velocity profiles, it was found that the Mean Pearson Correlation Coefficient is 0.77 (max = 0.90) and the Mean Intraclass Correlation Coefficient is 0.66 (max = 0.82). The densitometry analysis of osteoblast cells cultured on the designed microfluidic chip for four days revealed that the cell proliferation rate correlates positively with the measured WSS values. The proposed analysis can be applied to quantify the laminar flow in microfluidic chip experiments without specialized equipment.
Collapse
Affiliation(s)
- Hein Htet Aung
- Laboratory of Artificial Intelligence and Innovation in Medicine (AIIM), Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, 906 Kampangpetch 6 Rd., Talat Bang Khen, Lak Si, Bangkok 10210, Thailand
| | - Phattarin Pothipan
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Suwannabhumi Canal Rd, Bang Pla, Bang Phli District, Samut Prakan 10540, Thailand
| | - Jirasin Aswakool
- Laboratory of Artificial Intelligence and Innovation in Medicine (AIIM), Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, 906 Kampangpetch 6 Rd., Talat Bang Khen, Lak Si, Bangkok 10210, Thailand
| | - Siraphob Santironnarong
- Defence Technology Institute, Office of the Permanent Secretary of Defence (Chaengwattana) 7th Floor, 47/433 Moo 3, Ban Mai, Pak Kret, Nonthaburi 11120, Thailand
| | - Rungrueang Phatthanakun
- Synchrotron Light Research Institute, 111 University Avenue, Muang District, Nakhon Ratchasima 30000, Thailand
| | - Visarute Pinrod
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathumthani 12120, Thailand
| | - Thanakorn Jiemsakul
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathumthani 12120, Thailand
| | - Wares Chancharoen
- Laboratory of Artificial Intelligence and Innovation in Medicine (AIIM), Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, 906 Kampangpetch 6 Rd., Talat Bang Khen, Lak Si, Bangkok 10210, Thailand
| | - Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Suwannabhumi Canal Rd, Bang Pla, Bang Phli District, Samut Prakan 10540, Thailand
| |
Collapse
|
3
|
Dsouza C, Komarova SV. Mechanosensitivity and mechanotransductive properties of osteoclasts. Am J Physiol Cell Physiol 2024; 326:C95-C106. [PMID: 37982175 DOI: 10.1152/ajpcell.00347.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Mechanical loading is essential for maintaining bone health. Here, we aimed to investigate the role of ATP and ADP in the mechanotransduction of bone-resorptive osteoclasts. Single osteoclast in primary cultures from 10 to 12-wk-old mice was mechanically stimulated by a gentle touch with a micropipette. Changes in cytosolic free calcium [Ca2+]i were analyzed in Fura-2 loaded osteoclasts. The cell injury was assessed by analyzing the cellular Fura-2 loss and classified as severe or mild using k-means. Osteoclasts responded to mechanical stimuli with transient calcium elevation (primary responders) and transduced these signals to neighboring cells, which responded with delayed calcium elevations (secondary responders). Severely injured osteoclasts had higher calcium transients than mildly injured cells. Fluid shear stress similarly induced reversible cell injury in osteoclasts. Secondary responses were abolished by treatment with A-804598, a specific inhibitor of P2X7, but not suramin, a broad P2 receptor blocker. Osteoclasts responded to ATP and ADP with concentration-dependent changes in [Ca2+]i. We performed osteoclast micropipette stimulation in the presence of phosphoenolpyruvate and pyruvate kinase which converted all ADP in solution to ATP, or with hexokinase converting all ATP to ADP. Osteoclasts with mild membrane injury demonstrated similar calcium responses in ATP and ADP-rich environments. However, when the mechanotransductive signal to severe osteoclast injury was converted to ADP, the fraction of secondary responders and their [Ca2+]i amplitude was higher. This study suggests the importance of osteoclast mechanobiology and the role of ADP-mediated signaling in conditions of altered mechanical loading associated with bone loss.NEW & NOTEWORTHY Osteoclasts are rarely considered as cells that participate in mechanical signaling in bone. We show that osteoclasts are capable of sensing and transmitting mechanical signals to neighboring cells. Mechanical stimulation commonly induces minor repairable membrane injury in osteoclasts. ATP and especially ADP were found to play important roles in the mechanoresponsiveness of osteoclasts. This study highlights the importance of osteoclast mechanobiology especially in conditions of altered mechanical loading associated with bone loss, such as in microgravity.
Collapse
Affiliation(s)
- Chrisanne Dsouza
- Department of Experimental Surgery, McGill University, Montreal, Quebec, Canada
- Shriners Hospitals for Children-Canada, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Department of Experimental Surgery, McGill University, Montreal, Quebec, Canada
- Shriners Hospitals for Children-Canada, Montreal, Quebec, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Ma L, Wang W, Xu G, Li H, Liu F, Shao H, Zhang X, Ma Y, Li G, Li H, Gao S, Ling P. Connexin 43 in the function and homeostasis of osteocytes: a narrative review. ANNALS OF JOINT 2023; 9:10. [PMID: 38529291 PMCID: PMC10929443 DOI: 10.21037/aoj-23-65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/29/2023] [Indexed: 03/27/2024]
Abstract
Background and Objective Connexin 43 (Cx43) is the main gap junction (GJ) protein and hemichannel protein in bone tissue. It is involved in the formation of hemichannels and GJs and establishes channels that can communicate directly to exchange substances and signals, affecting the structure and function of osteocytes. CX43 is very important for the normal development of bone tissue and the establishment and balance of bone reconstruction. However, the molecular mechanisms by which CX43 regulates osteoblast function and homeostasis have been less well studied, and this article provides a review of research in this area. Methods We searched the PubMed, EMBASE, Cochrane Library, and Web of Science databases for studies published up to June 2023 using the keywords Connexin 43/Cx43 and Osteocytes. Screening of literatures according to inclusion and exclusion guidelines and summarized the results. Key Content and Findings Osteocytes, osteoblasts, and osteoclasts all express Cx43 and form an overall network through the interaction between GJs. Cx43 is not only involved in the mechanical response of bone tissue but also in the regulation of signal transduction, which could provide new molecular markers and novel targets for the treatment of certain bone diseases. Conclusions Cx43 is expressed in osteoblasts, osteoclasts, and osteoclasts and plays an important role in regulating the function, signal transduction, and mechanotransduction of osteocytes. This review offers a new contribution to the literature by summarizing the relationship between Cx43, a key protein of bone tissue, and osteoblasts.
Collapse
Affiliation(s)
- Liang Ma
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Post-doctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan, China
- Post-doctoral Station of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenzhao Wang
- Department of Orthopedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guixuan Xu
- Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- Department of Joint Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fei Liu
- Post-doctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan, China
| | - Huarong Shao
- Post-doctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan, China
| | - Xiuhua Zhang
- Post-doctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan, China
| | - Yuxia Ma
- Post-doctoral Station of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Gang Li
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hui Li
- Department of Operating Room, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuzhong Gao
- Post-doctoral Station of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peixue Ling
- Post-doctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan, China
| |
Collapse
|
5
|
LaGuardia JS, Shariati K, Bedar M, Ren X, Moghadam S, Huang KX, Chen W, Kang Y, Yamaguchi DT, Lee JC. Convergence of Calcium Channel Regulation and Mechanotransduction in Skeletal Regenerative Biomaterial Design. Adv Healthc Mater 2023; 12:e2301081. [PMID: 37380172 PMCID: PMC10615747 DOI: 10.1002/adhm.202301081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/20/2023] [Indexed: 06/30/2023]
Abstract
Cells are known to perceive their microenvironment through extracellular and intracellular mechanical signals. Upon sensing mechanical stimuli, cells can initiate various downstream signaling pathways that are vital to regulating proliferation, growth, and homeostasis. One such physiologic activity modulated by mechanical stimuli is osteogenic differentiation. The process of osteogenic mechanotransduction is regulated by numerous calcium ion channels-including channels coupled to cilia, mechanosensitive and voltage-sensitive channels, and channels associated with the endoplasmic reticulum. Evidence suggests these channels are implicated in osteogenic pathways such as the YAP/TAZ and canonical Wnt pathways. This review aims to describe the involvement of calcium channels in regulating osteogenic differentiation in response to mechanical loading and characterize the fashion in which those channels directly or indirectly mediate this process. The mechanotransduction pathway is a promising target for the development of regenerative materials for clinical applications due to its independence from exogenous growth factor supplementation. As such, also described are examples of osteogenic biomaterial strategies that involve the discussed calcium ion channels, calcium-dependent cellular structures, or calcium ion-regulating cellular features. Understanding the distinct ways calcium channels and signaling regulate these processes may uncover potential targets for advancing biomaterials with regenerative osteogenic capabilities.
Collapse
Affiliation(s)
- Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Dean T. Yamaguchi
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
6
|
Cui Y, Li C, Zeng X, Wei X, Li P, Cheng J, Xu Q, Yang Y. ATP purinergic receptor signalling promotes Sca-1 + cell proliferation and migration for vascular remodelling. Cell Commun Signal 2023; 21:173. [PMID: 37430253 PMCID: PMC10332060 DOI: 10.1186/s12964-023-01185-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/06/2023] [Indexed: 07/12/2023] Open
Abstract
AIMS Vascular resident stem cells expressing stem cell antigen-1 (Sca-1+ cells) promote vascular regeneration and remodelling following injury through migration, proliferation and differentiation. The aim of this study was to examine the contributions of ATP signalling through purinergic receptor type 2 (P2R) isoforms in promoting Sca-1+ cell migration and proliferation after vascular injury and to elucidate the main downstream signalling pathways. METHODS AND RESULTS ATP-evoked changes in isolated Sca-1+ cell migration were examined by transwell assays, proliferation by viable cell counting assays and intracellular Ca2+ signalling by fluorometry, while receptor subtype contributions and downstream signals were examined by pharmacological or genetic inhibition, immunofluorescence, Western blotting and quantitative RT-PCR. These mechanisms were further examined in mice harbouring TdTomato-labelled Sca-1+ cells with and without Sca-1+-targeted P2R knockout following femoral artery guidewire injury. Stimulation with ATP promoted cultured Sca-1+ cell migration, induced intracellular free calcium elevations primarily via P2Y2R stimulation and accelerated proliferation mainly via P2Y6R stimulation. Enhanced migration was inhibited by the ERK blocker PD98059 or P2Y2R-shRNA, while enhanced proliferation was inhibited by the P38 inhibitor SB203580. Femoral artery guidewire injury of the neointima increased the number of TdTomato-labelled Sca-1+ cells, neointimal area and the ratio of neointimal area to media area at 3 weeks post-injury, and all of these responses were reduced by P2Y2R knockdown. CONCLUSIONS ATP induces Sca-1+ cell migration through the P2Y2R-Ca2+-ERK signalling pathway, and enhances proliferation through the P2Y6R-P38-MAPK signalling pathway. Both pathways are essential for vascular remodelling following injury. Video Abstract.
Collapse
Affiliation(s)
- Yiqin Cui
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Chunshu Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Xinyi Zeng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Xiaoyu Wei
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Jun Cheng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China
| | - Qingbo Xu
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China.
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, 1-1 Xianglin Road, Luzhou, 646000, China.
| |
Collapse
|
7
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
8
|
Noronha-Matos JB, Pinto-Cardoso R, Bessa-Andrês C, Magalhães-Cardoso MT, Ferreirinha F, Costa MA, Marinhas J, Freitas R, Lemos R, Vilaça A, Oliveira A, Pelletier J, Sévigny J, Correia-de-Sá P. Silencing NTPDase3 activity rehabilitates the osteogenic commitment of post-menopausal stem cell bone progenitors. Stem Cell Res Ther 2023; 14:97. [PMID: 37076930 PMCID: PMC10116749 DOI: 10.1186/s13287-023-03315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Endogenously released adenine and uracil nucleotides favour the osteogenic commitment of bone marrow-derived mesenchymal stromal cells (BM-MSCs) through the activation of ATP-sensitive P2X7 and UDP-sensitive P2Y6 receptors. Yet, these nucleotides have their osteogenic potential compromised in post-menopausal (Pm) women due to overexpression of nucleotide metabolizing enzymes, namely NTPDase3. This prompted us to investigate whether NTPDase3 gene silencing or inhibition of its enzymatic activity could rehabilitate the osteogenic potential of Pm BM-MSCs. METHODS MSCs were harvested from the bone marrow of Pm women (69 ± 2 years old) and younger female controls (22 ± 4 years old). The cells were allowed to grow for 35 days in an osteogenic-inducing medium in either the absence or the presence of NTPDase3 inhibitors (PSB 06126 and hN3-B3s antibody); pre-treatment with a lentiviral short hairpin RNA (Lenti-shRNA) was used to silence the NTPDase3 gene expression. Immunofluorescence confocal microscopy was used to monitor protein cell densities. The osteogenic commitment of BM-MSCs was assessed by increases in the alkaline phosphatase (ALP) activity. The amount of the osteogenic transcription factor Osterix and the alizarin red-stained bone nodule formation. ATP was measured with the luciferin-luciferase bioluminescence assay. The kinetics of the extracellular ATP (100 µM) and UDP (100 µM) catabolism was assessed by HPLC RESULTS: The extracellular catabolism of ATP and UDP was faster in BM-MSCs from Pm women compared to younger females. The immunoreactivity against NTPDase3 increased 5.6-fold in BM-MSCs from Pm women vs. younger females. Selective inhibition or transient NTPDase3 gene silencing increased the extracellular accumulation of adenine and uracil nucleotides in cultured Pm BM-MSCs. Downregulation of NTPDase3 expression or activity rehabilitated the osteogenic commitment of Pm BM-MSCs measured as increases in ALP activity, Osterix protein cellular content and bone nodule formation; blockage of P2X7 and P2Y6 purinoceptors prevented this effect. CONCLUSIONS Data suggest that NTPDase3 overexpression in BM-MSCs may be a clinical surrogate of the osteogenic differentiation impairment in Pm women. Thus, besides P2X7 and P2Y6 receptors activation, targeting NTPDase3 may represent a novel therapeutic strategy to increase bone mass and reduce the osteoporotic risk of fractures in Pm women.
Collapse
Affiliation(s)
- José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal.
| | - Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Maria Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Maria Adelina Costa
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
- Departamento de Química, Instituto de Ciências Biomédicas Abel Salazar - Universidade Do Porto (ICBAS-UP), 4050-313, Porto, Portugal
| | - José Marinhas
- Serviço de Ortopedia e Traumatologia, Centro Hospitalar de Gaia - Espinho, 4434-502, Vila Nova de Gaia, Portugal
| | - Rolando Freitas
- Serviço de Ortopedia e Traumatologia, Centro Hospitalar de Gaia - Espinho, 4434-502, Vila Nova de Gaia, Portugal
| | - Rui Lemos
- Serviço de Ortopedia e Traumatologia, Centro Hospitalar de Gaia - Espinho, 4434-502, Vila Nova de Gaia, Portugal
| | - Adélio Vilaça
- Serviço de Ortopedia, Centro Hospitalar Universitário de Santo António, 4099-001, Porto, Portugal
| | - António Oliveira
- Serviço de Ortopedia, Centro Hospitalar Universitário de Santo António, 4099-001, Porto, Portugal
| | - Julie Pelletier
- Centre de Recherche en Rhumatologie et Immunologie, University Laval, 2325, rue de l'Université Québec, Québec, G1V 0A6, Canada
| | - Jean Sévigny
- Centre de Recherche en Rhumatologie et Immunologie, University Laval, 2325, rue de l'Université Québec, Québec, G1V 0A6, Canada
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal.
| |
Collapse
|
9
|
Zhao D, Wu J, Acosta FM, Xu H, Jiang JX. Connexin 43 hemichannels and prostaglandin E 2 release in anabolic function of the skeletal tissue to mechanical stimulation. Front Cell Dev Biol 2023; 11:1151838. [PMID: 37123401 PMCID: PMC10133519 DOI: 10.3389/fcell.2023.1151838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
Bone adapts to changes in the physical environment by modulating remodeling through bone resorption and formation to maintain optimal bone mass. As the most abundant connexin subtype in bone tissue, connexin 43 (Cx43)-forming hemichannels are highly responsive to mechanical stimulation by permitting the exchange of small molecules (<1.2 kDa) between bone cells and the extracellular environment. Upon mechanical stimulation, Cx43 hemichannels facilitate the release of prostaglandins E2 (PGE2), a vital bone anabolic factor from osteocytes. Although most bone cells are involved in mechanosensing, osteocytes are the principal mechanosensitive cells, and PGE2 biosynthesis is greatly enhanced by mechanical stimulation. Mechanical stimulation-induced PGE2 released from osteocytic Cx43 hemichannels acts as autocrine effects that promote β-catenin nuclear accumulation, Cx43 expression, gap junction function, and protects osteocytes against glucocorticoid-induced osteoporosis in cultured osteocytes. In vivo, Cx43 hemichannels with PGE2 release promote bone formation and anabolism in response to mechanical loading. This review summarizes current in vitro and in vivo understanding of Cx43 hemichannels and extracellular PGE2 release, and their roles in bone function and mechanical responses. Cx43 hemichannels could be a significant potential new therapeutic target for treating bone loss and osteoporosis.
Collapse
Affiliation(s)
- Dezhi Zhao
- School of Medicine, Northwest University, Xi’an, China
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Jiawei Wu
- School of Medicine, Northwest University, Xi’an, China
| | - Francisca M. Acosta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| |
Collapse
|
10
|
Huang H, He YM, Lin MM, Wang Y, Zhang X, Liang L, He X. P2X7Rs: new therapeutic targets for osteoporosis. Purinergic Signal 2023; 19:207-219. [PMID: 35106736 PMCID: PMC9984661 DOI: 10.1007/s11302-021-09836-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023] Open
Abstract
Increasing evidence suggests that both the occurrence and progression of osteoporosis are associated with inflammation, especially in primary osteoporosis. The maintenance of skeletal homeostasis is dependent on the complex regulation of bone metabolism. Numerous evidence suggested that purinoceptor networks are essential for bone homeostasis. In this review, the relationship between inflammation and the development of osteoporosis and the role of P2X7 receptor (P2X7R) in regulating the dynamic regulation of bone reconstruction were covered. We also discussed how P2X7R regulates the balance between resorption and bone formation by osteoblasts and reviewed the relevance of P2X7R polymorphisms in skeletal physiology. Finally, we analyzed potential targets of P2X7R for osteoporosis.
Collapse
Affiliation(s)
- Haoyun Huang
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yu-Mei He
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610041, China
| | - Miao-Miao Lin
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610041, China
| | - Yanchao Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaomei Zhang
- Laboratory Animal Center of Sichuan University, Chengdu, 610041, China
| | - Li Liang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xueling He
- Laboratory Animal Center of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
11
|
Shimizu N, Fujiwara K, Mayahara K, Motoyoshi M, Takahashi T. Tension force causes cell cycle arrest at G2/M phase in osteocyte-like cell line MLO-Y4. Heliyon 2023; 9:e13236. [PMID: 36798766 PMCID: PMC9925960 DOI: 10.1016/j.heliyon.2023.e13236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023] Open
Abstract
Bone remodelling is the process of bone resorption and formation, necessary to maintain bone structure or for adaptation to new conditions. Mechanical loadings, such as exercise, weight bearing and orthodontic force, play important roles in bone remodelling. During the remodelling process, osteocytes play crucial roles as mechanosensors to regulate osteoblasts and osteoclasts. However, the precise molecular mechanisms by which the mechanical stimuli affect the function of osteocytes remain unclear. In the present study, we analysed viability, cell cycle distribution and gene expression pattern of murine osteocyte-like MLO-Y4 cells exposed to tension force (TF). Cells were subjected to TF with 18% elongation at 6 cycles/min for 24 h using Flexcer Strain Unit (FX-3000). We found that TF stimulation induced cell cycle arrest at G2/M phase but not cell death in MLO-Y4 cells. Differentially expressed genes (DEGs) between TF-stimulated and unstimulated cells were identified by microarray analysis, and a marked increase in glutathione-S-transferase α (GSTA) family gene expression was observed in TF-stimulated cells. Enrichment analysis for the DEGs revealed that Gene Ontology (GO) terms and Kyoto Encyclopedia Genes and Genomes (KEGG) pathways related to the stress response were significantly enriched among the upregulated genes following TF. Consistent with these results, the production of reactive oxygen species (ROS) was elevated in TF-stimulated cells. Activation of the tumour suppressor p53, and upregulation of its downstream target GADD45A, were also observed in the stimulated cells. As GADD45A has been implicated in the promotion of G2/M cell cycle arrest, these observations may suggest that TF stress leads to G2/M arrest at least in part in a p53-dependent manner.
Collapse
Affiliation(s)
- Natsuo Shimizu
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Applied Oral Science, Nihon University Graduate School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kyoko Fujiwara
- Department of Anatomy, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Corresponding author. Department of Anatomy, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.
| | - Kotoe Mayahara
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Clinical Research, Dental Research Centre, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Mitsuru Motoyoshi
- Department of Orthodontics, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Clinical Research, Dental Research Centre, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Tomihisa Takahashi
- Department of Anatomy, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
- Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, 1-8-3 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| |
Collapse
|
12
|
Reyes Fernandez PC, Wright CS, Masterson AN, Yi X, Tellman TV, Bonteanu A, Rust K, Noonan ML, White KE, Lewis KJ, Sankar U, Hum JM, Bix G, Wu D, Robling AG, Sardar R, Farach-Carson MC, Thompson WR. Gabapentin Disrupts Binding of Perlecan to the α 2δ 1 Voltage Sensitive Calcium Channel Subunit and Impairs Skeletal Mechanosensation. Biomolecules 2022; 12:biom12121857. [PMID: 36551284 PMCID: PMC9776037 DOI: 10.3390/biom12121857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Our understanding of how osteocytes, the principal mechanosensors within bone, sense and perceive force remains unclear. Previous work identified "tethering elements" (TEs) spanning the pericellular space of osteocytes and transmitting mechanical information into biochemical signals. While we identified the heparan sulfate proteoglycan perlecan (PLN) as a component of these TEs, PLN must attach to the cell surface to induce biochemical responses. As voltage-sensitive calcium channels (VSCCs) are critical for bone mechanotransduction, we hypothesized that PLN binds the extracellular α2δ1 subunit of VSCCs to couple the bone matrix to the osteocyte membrane. Here, we showed co-localization of PLN and α2δ1 along osteocyte dendritic processes. Additionally, we quantified the molecular interactions between α2δ1 and PLN domains and demonstrated for the first time that α2δ1 strongly associates with PLN via its domain III. Furthermore, α2δ1 is the binding site for the commonly used pain drug, gabapentin (GBP), which is associated with adverse skeletal effects when used chronically. We found that GBP disrupts PLN::α2δ1 binding in vitro, and GBP treatment in vivo results in impaired bone mechanosensation. Our work identified a novel mechanosensory complex within osteocytes composed of PLN and α2δ1, necessary for bone force transmission and sensitive to the drug GBP.
Collapse
Affiliation(s)
- Perla C. Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Christian S. Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Adrianna N. Masterson
- Department of Chemistry and Chemical Biology, School of Science, Indiana University, Indianapolis, IN 46202, USA
| | - Xin Yi
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Tristen V. Tellman
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Andrei Bonteanu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - Katie Rust
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
| | - Megan L. Noonan
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Kenneth E. White
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Karl J. Lewis
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Uma Sankar
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Julia M. Hum
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA
| | - Gregory Bix
- Departments of Neurosurgery and Neurology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - Alexander G. Robling
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Rajesh Sardar
- Department of Chemistry and Chemical Biology, School of Science, Indiana University, Indianapolis, IN 46202, USA
| | - Mary C. Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - William R. Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Division of Biomedical Science, College of Osteopathic Medicine, Marian University, Indianapolis, IN 46222, USA
- Correspondence:
| |
Collapse
|
13
|
Limraksasin P, Nattasit P, Manokawinchoke J, Tiskratok W, Vinaikosol N, Okawa H, Limjeerajarus CN, Limjeerajarus N, Pavasant P, Osathanon T, Egusa H. Application of shear stress for enhanced osteogenic differentiation of mouse induced pluripotent stem cells. Sci Rep 2022; 12:19021. [PMID: 36347883 PMCID: PMC9643422 DOI: 10.1038/s41598-022-21479-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/27/2022] [Indexed: 11/10/2022] Open
Abstract
The self-organizing potential of induced pluripotent stem cells (iPSCs) represents a promising tool for bone tissue engineering. Shear stress promotes the osteogenic differentiation of mesenchymal stem cells, leading us to hypothesize that specific shear stress could enhance the osteogenic differentiation of iPSCs. For osteogenesis, embryoid bodies were formed for two days and then maintained in medium supplemented with retinoic acid for three days, followed by adherent culture in osteogenic induction medium for one day. The cells were then subjected to shear loading (0.15, 0.5, or 1.5 Pa) for two days. Among different magnitudes tested, 0.5 Pa induced the highest levels of osteogenic gene expression and greatest mineral deposition, corresponding to upregulated connexin 43 (Cx43) and phosphorylated Erk1/2 expression. Erk1/2 inhibition during shear loading resulted in decreased osteogenic gene expression and the suppression of mineral deposition. These results suggest that shear stress (0.5 Pa) enhances the osteogenic differentiation of iPSCs, partly through Cx43 and Erk1/2 signaling. Our findings shed light on the application of shear-stress technology to improve iPSC-based tissue-engineered bone for regenerative bone therapy.
Collapse
Affiliation(s)
- Phoonsuk Limraksasin
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan ,grid.7922.e0000 0001 0244 7875Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Rd. Pathumwan, Bangkok, 10330 Thailand
| | - Praphawi Nattasit
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Jeeranan Manokawinchoke
- grid.7922.e0000 0001 0244 7875Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Watcharaphol Tiskratok
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Naruephorn Vinaikosol
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Hiroko Okawa
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Chalida Nakalekha Limjeerajarus
- grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Nuttapol Limjeerajarus
- grid.7922.e0000 0001 0244 7875Office of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand ,grid.512238.f0000 0004 0625 2348Research Center for Advanced Energy Technology, Faculty of Engineering, Thai-Nichi Institute of Technology, Bangkok, Thailand
| | - Prasit Pavasant
- grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Rd. Pathumwan, Bangkok, 10330 Thailand
| | - Thanaphum Osathanon
- grid.7922.e0000 0001 0244 7875Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Rd. Pathumwan, Bangkok, 10330 Thailand
| | - Hiroshi Egusa
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan ,grid.69566.3a0000 0001 2248 6943Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai-city, 980-8575 Japan
| |
Collapse
|
14
|
Chen L, Hu B, Wang X, Chen Y, Zhou B. Functional role of cyanidin-3-O-glucoside in osteogenesis: A pilot study based on RNA-seq analysis. Front Nutr 2022; 9:995643. [PMID: 36245484 PMCID: PMC9562617 DOI: 10.3389/fnut.2022.995643] [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: 07/16/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cyanidin-3-O-glucoside (C3G) is the most widely distributed anthocyanin and it can reportedly reduce the risk of osteoporosis, but the molecular mechanism by which C3G promotes bone formation is poorly understood. In the current study, RNA sequencing (RNA-seq) was used to investigate the mechanism of action of C3G in osteogenesis. MC3T3-E1 mouse osteoblasts were divided into a C3G (100 μmol/L)-treated group and a vehicle-treated control group, and differentially expressed genes (DEGs) in groups were evaluated via RNA-seq analysis. The functions of the DEGs were evaluated by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and the genes were validated by quantitative real-time PCR. The RNA-seq analysis identified 34 genes that were upregulated in C3G-treated cells compared to vehicle-treated cells, and 17 that were downregulated GO and KEGG pathway analyses indicated that these genes were highly enriched in functions related to lysosomes and glycolipid biosynthesis, among others. The differential expression of ATPase H+-transporting V0 subunit C (Atp6v0c), chemokine (C-X3-C motif) ligand 1 (Cx3cl1), and lymphocyte antigen 6 complex, locus A (Ly6a) genes was validated by quantitative real-time-PCR. Because these genes have been previously implicated in osteoporosis, they are potential target genes of C3G action in MC3T3-E1 cells. These results provide molecular level evidence for the therapeutic potential of C3G in the treatment of osteoporosis and other disorders of bone metabolism.
Collapse
Affiliation(s)
- Lin Chen
- School of Public Health, Shenyang Medical College, Shenyang, China
| | - Bosen Hu
- School of Public Health, Shenyang Medical College, Shenyang, China
| | - Xiaohong Wang
- School of Public Health, Shenyang Medical College, Shenyang, China
| | - Yong Chen
- Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Bo Zhou
- School of Public Health, Shenyang Medical College, Shenyang, China
- *Correspondence: Bo Zhou
| |
Collapse
|
15
|
Impact of Fluid Flow Shear Stress on Osteoblast Differentiation and Cross-Talk with Articular Chondrocytes. Int J Mol Sci 2022; 23:ijms23169505. [PMID: 36012760 PMCID: PMC9408926 DOI: 10.3390/ijms23169505] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Bone cells, in particular osteoblasts, are capable of communication with each other during bone growth and homeostasis. More recently it has become clear that they also communicate with other cell-types; including chondrocytes in articular cartilage. One way that this process is facilitated is by interstitial fluid movement within the pericellular and extracellular matrices. This stimulus is also an important mechanical signal in skeletal tissues, and is known to generate shear stresses at the micron-scale (known as fluid flow shear stresses (FFSS)). The primary aim of this study was to develop and characterize an in vitro bone–cartilage crosstalk system, to examine the effect of FFSS on these cell types. Specifically, we evaluated the response of osteoblasts and chondrocytes to FFSS and the effect of FFSS-induced soluble factors from the former, on the latter. This system will ultimately be used to help us understand the role of subchondral bone damage in articular cartilage degeneration. We also carried out a comparison of responses between cell lines and primary murine cells in this work. Our findings demonstrate that primary cells produce a more reliable and reproducible response to FFSS. Furthermore we found that at lower magnitudes , direct FFSS produces anabolic responses in both chondrocytes and osteoblasts, whereas higher levels produce more catabolic responses. Finally we show that exposure to osteoblast-derived factors in conditioned media experiments produced similarly catabolic changes in primary chondrocytes.
Collapse
|
16
|
Dsouza C, Moussa MS, Mikolajewicz N, Komarova SV. Extracellular ATP and its derivatives provide spatiotemporal guidance for bone adaptation to wide spectrum of physical forces. Bone Rep 2022; 17:101608. [PMID: 35992507 PMCID: PMC9385560 DOI: 10.1016/j.bonr.2022.101608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
ATP is a ubiquitous intracellular molecule critical for cellular bioenergetics. ATP is released in response to mechanical stimulation through vesicular release, small tears in cellular plasma membranes, or when cells are destroyed by traumatic forces. Extracellular ATP is degraded by ecto-ATPases to form ADP and eventually adenosine. ATP, ADP, and adenosine signal through purinergic receptors, including seven P2X ATP-gated cation channels, seven G-protein coupled P2Y receptors responsive to ATP and ADP, and four P1 receptors stimulated by adenosine. The goal of this review is to build a conceptual model of the role of different components of this complex system in coordinating cellular responses that are appropriate to the degree of mechanical stimulation, cell proximity to the location of mechanical injury, and time from the event. We propose that route and amount of ATP release depend on the scale of mechanical forces, ranging from vesicular release of small ATP boluses upon membrane deformation, to leakage of ATP through resealable plasma membrane tears, to spillage of cellular content due to destructive forces. Correspondingly, different P2 receptors responsive to ATP will be activated according to their affinity at the site of mechanical stimulation. ATP is a small molecule that readily diffuses through the environment, bringing the signal to the surrounding cells. ATP is also degraded to ADP which can stimulate a distinct set of P2 receptors. We propose that depending on the magnitude of mechanical forces and distance from the site of their application, ATP/ADP profiles will be different, allowing the relay of information about tissue level injury and proximity. Lastly, ADP is degraded to adenosine acting via its P1 receptors. The presence of large amounts of adenosine without ATP, indicates that an active source of ATP release is no longer present, initiating the transition to the recovery phase. This model consolidates the knowledge regarding the individual components of the purinergic system into a conceptual framework of choreographed responses to physical forces. Cellular bioenergetic molecule ATP is released when cell is mechanically stimulated. ATP release is proportional to the amount of cellular damage. ATP diffusion and transformation to ADP indicates the proximity to the damage. Purinergic receptors form a network choreographing cell response to physical forces. Complete transformation of ATP to adenosine initiates the recovery phase.
Collapse
Affiliation(s)
- Chrisanne Dsouza
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
| | - Mahmoud S. Moussa
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Nicholas Mikolajewicz
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Svetlana V. Komarova
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
- Corresponding author.
| |
Collapse
|
17
|
Esposito L, Minutolo V, Gargiulo P, Fraldi M. Symmetry breaking and effects of nutrient walkway in time-dependent bone remodeling incorporating poroelasticity. Biomech Model Mechanobiol 2022; 21:999-1020. [PMID: 35394267 PMCID: PMC9132879 DOI: 10.1007/s10237-022-01573-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/07/2022] [Indexed: 12/03/2022]
Abstract
Bone is an extraordinary biological material that continuously adapts its hierarchical microstructure to respond to static and dynamic loads for offering optimal mechanical features, in terms of stiffness and toughness, across different scales, from the sub-microscopic constituents within osteons—where the cyclic activity of osteoblasts, osteoclasts, and osteocytes redesigns shape and percentage of mineral crystals and collagen fibers—up to the macroscopic level, with growth and remodeling processes that modify the architecture of both compact and porous bone districts. Despite the intrinsic complexity of the bone mechanobiology, involving coupling phenomena of micro-damage, nutrients supply driven by fluid flowing throughout hierarchical networks, and cells turnover, successful models and numerical algorithms have been presented in the literature to predict, at the macroscale, how bone remodels under mechanical stimuli, a fundamental issue in many medical applications such as optimization of femur prostheses and diagnosis of the risk fracture. Within this framework, one of the most classical strategies employed in the studies is the so-called Stanford’s law, which allows uploading the effect of the time-dependent load-induced stress stimulus into a biomechanical model to guess the bone structure evolution. In the present work, we generalize this approach by introducing the bone poroelasticity, thus incorporating in the model the role of the fluid content that, by driving nutrients and contributing to the removal of wastes of bone tissue cells, synergistically interacts with the classical stress fields to change homeostasis states, local saturation conditions, and reorients the bone density rate, in this way affecting growth and remodeling. Through two paradigmatic example applications, i.e. a cylindrical slice with internal prescribed displacements idealizing a tract of femoral diaphysis pushed out by the pressure exerted by a femur prosthesis and a bone element in a form of a bent beam, it is highlighted that the present model is capable to catch more realistically both the transition between spongy and cortical regions and the expected non-symmetrical evolution of bone tissue density in the medium–long term, unpredictable with the standard approach. A real study case of a femur is also considered at the end in order to show the effectiveness of the proposed remodeling algorithm.
Collapse
Affiliation(s)
- L Esposito
- Department Engineering, University of Campania "Luigi Vanvitelli", Aversa, Italy
| | - V Minutolo
- Department Engineering, University of Campania "Luigi Vanvitelli", Aversa, Italy
| | - P Gargiulo
- Institute for Biomedical and Neural Engineering, Reykjavík University, Reykjavík, Iceland
- Department of Science, Landspítali Hospital, Reykjavík, Iceland
| | - M Fraldi
- Department of Structures for Engineering and Architecture, University of Napoli "Federico II", Napoli, Italy.
| |
Collapse
|
18
|
Gould NR, Leser JM, Torre OM, Khairallah RJ, Ward CW, Stains JP. In vitro Fluid Shear Stress Induced Sclerostin Degradation and CaMKII Activation in Osteocytes. Bio Protoc 2021; 11:e4251. [PMID: 35005095 PMCID: PMC8678913 DOI: 10.21769/bioprotoc.4251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/18/2021] [Accepted: 09/27/2021] [Indexed: 11/19/2023] Open
Abstract
Bone is a dynamic tissue that adapts to changes in its mechanical environment. Mechanical stimuli pressurize interstitial fluid in the lacunar-canalicular system within the bone matrix, causing fluid shear stress (FSS) across bone embedded, mechano-sensitive osteocytes. Therefore, modeling this mechanical stimulus in vitro is vital for identifying mechano-transduction cascades that contribute to the regulation of mechano-responsive proteins, such as the Wnt/β-catenin antagonist, sclerostin, which is reduced in response to FSS. Recently, we reported the rapid post-translational degradation of sclerostin protein in bone cells following FSS. Given the fundamental nature of sclerostin to bone physiology and the nuances of studying its rapid post-translational control, here, we detail our FSS protocol, and adaptations that can be made, to stimulate Ocy454 osteocyte-like cells to study sclerostin protein in vitro. While this protocol is optimized for detecting sclerostin degradation by western blot, this protocol can be adapted to examine transcriptional changes with RT-qPCR, cellular dynamics with live cell imaging, or secreted factors in the FSS buffer. This protocol utilizes 3D-printed FSS tips that are compatible with commercially available 96-well plates, allowing for high experimental accessibility, versatility, and throughput. However, this protocol can be adapted for any FSS chamber. It can also be combined with pharmacological inhibitors or genetic manipulations to interrogate the role of specific cellular components. In all, this experimental set-up and protocol is highly adaptable to allow for many experimental outcomes to examine many aspects of cell mechano-transduction.
Collapse
Affiliation(s)
- Nicole R. Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | - Jenna M. Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | - Olivia M. Torre
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | | | - Christopher W. Ward
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| | - Joseph P. Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
19
|
Tattersall L, Shah KM, Lath DL, Singh A, Down JM, De Marchi E, Williamson A, Di Virgilio F, Heymann D, Adinolfi E, Fraser WD, Green D, Lawson MA, Gartland A. The P2RX7B splice variant modulates osteosarcoma cell behaviour and metastatic properties. J Bone Oncol 2021; 31:100398. [PMID: 35340569 PMCID: PMC8948168 DOI: 10.1016/j.jbo.2021.100398] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
P2RX7B expression confers a survival advantage in TE85+P2RX7B and MNNG-HOS+P2RX7B OS cell lines. P2RX7B expression reduced cell adhesion and activation promoted invasion and migration in vitro. MNNG-HOS+P2RX7B tumours in vivo exhibited ectopic bone formation that A740003 reduced. Expression of P2RX7B in primary tumour cells increased the propensity to metastasise to the lungs. A novel gene axis, FN1/LOX/PDGFB/IGFBP3/BMP4 was downregulated in response to A740003.
Background Osteosarcoma (OS) is the most common type of primary bone cancer affecting children and adolescents. OS has a high propensity to spread meaning the disease is often incurable and fatal. There have been no improvements in survival rates for decades. This highlights an urgent need for the development of novel therapeutic strategies. Here, we report in vitro and in vivo data that demonstrates the role of purinergic signalling, specifically, the B isoform of the purinergic receptor P2RX7 (P2RX7B), in OS progression and metastasis. Methods TE85 and MNNG-HOS OS cells were transfected with P2RX7B. These cell lines were then characterised and assessed for proliferation, cell adhesion, migration and invasion in vitro. We used these cells to perform both paratibial and tail vein injected mouse studies where the primary tumour, bone and lungs were analysed. We used RNA-seq to identify responsive pathways relating to P2RX7B. Results Our data shows that P2RX7B expression confers a survival advantage in TE85 + P2RX7B and MNNG-HOS + P2RX7B human OS cell lines in vitro that is minimised following treatment with A740003, a specific P2RX7 antagonist. P2RX7B expression reduced cell adhesion and P2RX7B activation promoted invasion and migration in vitro, demonstrating a metastatic phenotype. Using an in vivo OS xenograft model, MNNG-HOS + P2RX7B tumours exhibited cancer-associated ectopic bone formation that was abrogated with A740003 treatment. A pro-metastatic phenotype was further demonstrated in vivo as expression of P2RX7B in primary tumour cells increased the propensity of tumour cells to metastasise to the lungs. RNA-seq identified a novel gene axis, FN1/LOX/PDGFB/IGFBP3/BMP4, downregulated in response to A740003 treatment. Conclusion Our data illustrates a role for P2RX7B in OS tumour growth, progression and metastasis. We show that P2RX7B is a future therapeutic target in human OS.
Collapse
Affiliation(s)
- Luke Tattersall
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, UK
| | - Karan M. Shah
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, UK
| | - Darren L. Lath
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, UK
| | - Archana Singh
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Jennifer M. Down
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, UK
| | - Elena De Marchi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Italy
| | - Alex Williamson
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, UK
| | - Francesco Di Virgilio
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Italy
| | - Dominique Heymann
- Université de Nantes, Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Elena Adinolfi
- Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, Italy
| | - William D. Fraser
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
- Clinical Biochemistry, Norfolk and Norwich University Hospital, Norwich Research Park, Norwich, UK
| | - Darrell Green
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Michelle A. Lawson
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, UK
| | - Alison Gartland
- The Mellanby Centre for Musculoskeletal Research, Department of Oncology and Metabolism, The University of Sheffield, UK
- Corresponding author.
| |
Collapse
|
20
|
Carter A, Popowski K, Cheng K, Greenbaum A, Ligler FS, Moatti A. Enhancement of Bone Regeneration Through the Converse Piezoelectric Effect, A Novel Approach for Applying Mechanical Stimulation. Bioelectricity 2021; 3:255-271. [PMID: 35018335 PMCID: PMC8742263 DOI: 10.1089/bioe.2021.0019] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Serious bone injuries have devastating effects on the lives of patients including limiting working ability and high cost. Orthopedic implants can aid in healing injuries to an extent that exceeds the natural regenerative capabilities of bone to repair fractures or large bone defects. Autografts and allografts are the standard implants used, but disadvantages such as donor site complications, a limited quantity of transplantable bone, and high costs have led to an increased demand for synthetic bone graft substitutes. However, replicating the complex physiological properties of biological bone, much less recapitulating its complex tissue functions, is challenging. Extensive efforts to design biocompatible implants that mimic the natural healing processes in bone have led to the investigation of piezoelectric smart materials because the bone has natural piezoelectric properties. Piezoelectric materials facilitate bone regeneration either by accumulating electric charge in response to mechanical stress, which mimics bioelectric signals through the direct piezoelectric effect or by providing mechanical stimulation in response to electrical stimulation through the converse piezoelectric effect. Although both effects are beneficial, the converse piezoelectric effect can address bone atrophy from stress shielding and immobility by improving the mechanical response of a healing defect. Mechanical stimulation has a positive impact on bone regeneration by activating cellular pathways that increase bone formation and decrease bone resorption. This review will highlight the potential of the converse piezoelectric effect to enhance bone regeneration by discussing the activation of beneficial cellular pathways, the properties of piezoelectric biomaterials, and the potential for the more effective administration of the converse piezoelectric effect using wireless control.
Collapse
Affiliation(s)
- Amber Carter
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Kristen Popowski
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Ke Cheng
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Alon Greenbaum
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Adele Moatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
21
|
Trompeter N, Gardinier JD, DeBarros V, Boggs M, Gangadharan V, Cain WJ, Hurd L, Duncan RL. Insulin-like growth factor-1 regulates the mechanosensitivity of chondrocytes by modulating TRPV4. Cell Calcium 2021; 99:102467. [PMID: 34530313 PMCID: PMC8541913 DOI: 10.1016/j.ceca.2021.102467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Both mechanical and biochemical stimulation are required for maintaining the integrity of articular cartilage. However, chondrocytes respond differently to mechanical stimuli in osteoarthritic cartilage when biochemical signaling pathways, such as Insulin-like Growth Factor-1 (IGF-1), are altered. The Transient Receptor Potential Vanilloid 4 (TRPV4) channel is central to chondrocyte mechanotransduction and regulation of cartilage homeostasis. Here, we propose that changes in IGF-1 can modulate TRPV4 channel activity. We demonstrate that physiologic levels of IGF-1 suppress hypotonic-induced TRPV4 currents and intracellular calcium flux by increasing apparent cell stiffness that correlates with actin stress fiber formation. Disruption of F-actin following IGF-1 treatment results in the return of the intracellular calcium response to hypotonic swelling. Using point mutations of the TRPV4 channel at the microtubule-associated protein 7 (MAP-7) site shows that regulation of TRPV4 by actin is mediated via the interaction of actin with the MAP-7 domain of TRPV4. We further highlight that ATP release, a down-stream response to mechanical stimulation in chondrocytes, is mediated by TRPV4 during hypotonic challenge. This response is significantly abrogated with IGF-1 treatment. As chondrocyte mechanosensitivity is greatly altered during osteoarthritis progression, IGF-1 presents as a promising candidate for prevention and treatment of articular cartilage damage.
Collapse
Affiliation(s)
- Nicholas Trompeter
- Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Joseph D Gardinier
- Biomechanics and Movement Science Program, University of Delaware, Newark, DE, United States; Bone and Joint Center, Henry Ford Hospital, Detroit, MI, United States
| | - Victor DeBarros
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Mary Boggs
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Vimal Gangadharan
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - William J Cain
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Lauren Hurd
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Randall L Duncan
- Biomedical Engineering, University of Delaware, Newark, DE, United States; Biomechanics and Movement Science Program, University of Delaware, Newark, DE, United States; Department of Biological Sciences, University of Delaware, Newark, DE, United States; Department of Biology, University of Michigan-Flint, Flint, MI, United States.
| |
Collapse
|
22
|
The mechanosensory and mechanotransductive processes mediated by ion channels and the impact on bone metabolism: A systematic review. Arch Biochem Biophys 2021; 711:109020. [PMID: 34461086 DOI: 10.1016/j.abb.2021.109020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Mechanical environments were associated with alterations in bone metabolism. Ion channels present on bone cells are indispensable for bone metabolism and can be directly or indirectly activated by mechanical stimulation. This review aimed to discuss the literature reporting the mechanical regulatory effects of ion channels on bone cells and bone tissue. An electronic search was conducted in PubMed, Embase and Web of Science. Studies about mechanically induced alteration of bone cells and bone tissue by ion channels were included. Ion channels including TRP family channels, Ca2+ release-activated Ca2+ channels (CRACs), Piezo1/2 channels, purinergic receptors, NMDA receptors, voltage-sensitive calcium channels (VSCCs), TREK2 potassium channels, calcium- and voltage-dependent big conductance potassium (BKCa) channels, small conductance, calcium-activated potassium (SKCa) channels and epithelial sodium channels (ENaCs) present on bone cells and bone tissue participate in the mechanical regulation of bone development in addition to contributing to direct or indirect mechanotransduction such as altered membrane potential and ionic flux. Physiological (beneficial) mechanical stimulation could induce the anabolism of bone cells and bone tissue through ion channels, but abnormal (harmful) mechanical stimulation could also induce the catabolism of bone cells and bone tissue through ion channels. Functional expression of ion channels is vital for the mechanotransduction of bone cells. Mechanical activation (opening) of ion channels triggers ion influx and induces the activation of intracellular modulators that can influence bone metabolism. Therefore, mechanosensitive ion channels provide new insights into therapeutic targets for the treatment of bone-related diseases such as osteopenia and aseptic implant loosening.
Collapse
|
23
|
Atif AR, Pujari-Palmer M, Tenje M, Mestres G. A microfluidics-based method for culturing osteoblasts on biomimetic hydroxyapatite. Acta Biomater 2021; 127:327-337. [PMID: 33785452 DOI: 10.1016/j.actbio.2021.03.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/02/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
The reliability of conventional cell culture studies to evaluate biomaterials is often questioned, as in vitro outcomes may contradict results obtained through in vivo assays. Microfluidics technology has the potential to reproduce complex physiological conditions by allowing for fine control of microscale features such as cell confinement and flow rate. Having a continuous flow during cell culture is especially advantageous for bioactive biomaterials such as calcium-deficient hydroxyapatite (HA), which may otherwise alter medium composition and jeopardize cell viability, potentially producing false negative results in vitro. In this work, HA was integrated into a microfluidics-based platform (HA-on-chip) and the effect of varied flow rates (2, 8 and 14 µl/min, corresponding to 0.002, 0.008 and 0.014 dyn/cm2, respectively) was evaluated. A HA sample placed in a well plate (HA-static) was included as a control. While substantial calcium depletion and phosphate release occurred in static conditions, the concentration of ions in HA-on-chip samples remained similar to those of fresh medium, particularly at higher flow rates. Pre-osteoblast-like cells (MC3T3-E1) exhibited a significantly higher degree of proliferation on HA-on-chip (8 μl/min flow rate) as compared to HA-static. However, cell differentiation, analysed by alkaline phosphatase (ALP) activity, showed low values in both conditions. This study indicates that cells respond differently when cultured on HA under flow compared to static conditions, which indicates the need for more physiologically relevant methods to increase the predictive value of in vitro studies used to evaluate biomaterials. STATEMENT OF SIGNIFICANCE: There is a lack of correlation between the results obtained when testing some biomaterials under cell culture as opposed to animal models. To address this issue, a cell culture method with slightly enhanced physiological relevance was developed by incorporating a biomaterial, known to regenerate bone, inside of a microfluidic platform that enabled a continuous supply of cell culture medium. Since the utilized biomaterial interacts with surrounding ions, the perfusion of medium allowed for shielding of these changes similarly as would happen in the body. The experimental outcomes observed in the dynamic platform were different than those obtained with standard static cell culture systems, proving the key role of the platform in the assessment of biomaterials.
Collapse
Affiliation(s)
- Abdul Raouf Atif
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| | - Michael Pujari-Palmer
- Division of Applied Materials Science, Department of Materials Science and Engineering, Uppsala University, 751 22 Uppsala, Sweden
| | - Maria Tenje
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden
| | - Gemma Mestres
- Division of Microsystems Technology, Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, 751 22 Uppsala, Sweden.
| |
Collapse
|
24
|
Liu H, Usprech JF, Parameshwar PK, Sun Y, Simmons CA. Combinatorial screen of dynamic mechanical stimuli for predictive control of MSC mechano-responsiveness. SCIENCE ADVANCES 2021; 7:7/19/eabe7204. [PMID: 33962940 PMCID: PMC8104874 DOI: 10.1126/sciadv.abe7204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/19/2021] [Indexed: 05/05/2023]
Abstract
Mechanobiological-based control of mesenchymal stromal cells (MSCs) to facilitate engineering and regeneration of load-bearing tissues requires systematic investigations of specific dynamic mechanical stimulation protocols. Using deformable membrane microdevice arrays paired with combinatorial experimental design and modeling, we probed the individual and integrative effects of mechanical stimulation parameters (strain magnitude, rate at which strain is changed, and duty period) on myofibrogenesis and matrix production of MSCs in three-dimensional hydrogels. These functions were found to be dominantly influenced by a previously unidentified, higher-order interactive effect between strain magnitude and duty period. Empirical models based on our combinatorial cue-response data predicted an optimal loading regime in which strain magnitude and duty period were increased synchronously over time, which was validated to most effectively promote MSC matrix production. These findings inform the design of loading regimes for MSC-based engineered tissues and validate a broadly applicable approach to probe multifactorial regulating effects of mechanobiological cues.
Collapse
Affiliation(s)
- Haijiao Liu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Jenna F Usprech
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Prabu Karthick Parameshwar
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
25
|
Wright CS, Robling AG, Farach-Carson MC, Thompson WR. Skeletal Functions of Voltage Sensitive Calcium Channels. Curr Osteoporos Rep 2021; 19:206-221. [PMID: 33721180 PMCID: PMC8216424 DOI: 10.1007/s11914-020-00647-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/15/2022]
Abstract
Voltage-sensitive calcium channels (VSCCs) are ubiquitous multimeric protein complexes that are necessary for the regulation of numerous physiological processes. VSCCs regulate calcium influx and various intracellular processes including muscle contraction, neurotransmission, hormone secretion, and gene transcription, with function specificity defined by the channel's subunits and tissue location. The functions of VSCCs in bone are often overlooked since bone is not considered an electrically excitable tissue. However, skeletal homeostasis and adaptation relies heavily on VSCCs. Inhibition or deletion of VSCCs decreases osteogenesis, impairs skeletal structure, and impedes anabolic responses to mechanical loading. RECENT FINDINGS: While the functions of VSCCs in osteoclasts are less clear, VSCCs have distinct but complementary functions in osteoblasts and osteocytes. PURPOSE OF REVIEW: This review details the structure, function, and nomenclature of VSCCs, followed by a comprehensive description of the known functions of VSCCs in bone cells and their regulation of bone development, bone formation, and mechanotransduction.
Collapse
Affiliation(s)
- Christian S Wright
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic & Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77054, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
26
|
Purinergic Signaling Mediates PTH and Fluid Flow-Induced Osteoblast Proliferation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6674570. [PMID: 33575337 PMCID: PMC7864748 DOI: 10.1155/2021/6674570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/09/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023]
Abstract
Both parathyroid hormone (PTH) and mechanical signals are able to regulate bone growth and regeneration. They also can work synergistically to regulate osteoblast proliferation, but little is known about the mechanisms how PTH and mechanical signals interact with each other during this process. In this study, we investigated responses of MC3T3-E1 osteoblasts to PTH and oscillatory fluid flow. We found that osteoblasts are more sensitive to mechanical signals in the presence of PTH according to ERK1/2 phosphorylation, ATP release, CREB phosphorylation, and cell proliferation. PTH may also reduce the osteoblast refractory period after desensitization due to mechanical signals. We further found that the synergistic responses of osteoblasts to fluid flow or ATP with PTH had similar patterns, suggesting that synergy between fluid flow and PTH may be through the ATP pathway. After we inhibited ATP effects using apyrase in osteoblasts, their synergistic responses to mechanical stimulation and PTH were also inhibited. Additionally, knocking down P2Y2 purinergic receptors can significantly attenuate osteoblast synergistic responses to mechanical stimulation and PTH in terms of ERK1/2 phosphorylation, CREB phosphorylation, and cell proliferation. Thus, our results suggest that PTH enhances mechanosensitivity of osteoblasts via a mechanism involving ATP and P2Y2 purinergic receptors.
Collapse
|
27
|
Du J, Yang J, He Z, Cui J, Yang Y, Xu M, Qu X, Zhao N, Yan M, Li H, Yu Z. Osteoblast and Osteoclast Activity Affect Bone Remodeling Upon Regulation by Mechanical Loading-Induced Leukemia Inhibitory Factor Expression in Osteocytes. Front Mol Biosci 2020; 7:585056. [PMID: 33324677 PMCID: PMC7726425 DOI: 10.3389/fmolb.2020.585056] [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: 07/19/2020] [Accepted: 10/22/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose Bone remodeling is affected by mechanical stimulation. Osteocytes are the primary mechanical load-sensing cells in the bone, and can regulate osteoblast and osteoclast activity, thus playing a key role in bone remodeling. Further, bone mass during exercise is also regulated by Leukemia inhibitory factor (LIF). This study aimed to investigate the role of LIF in the mechanical response of the bone, in vivo and in vitro, and to elucidate the mechanism by which osteocytes secrete LIF to regulate osteoblasts and osteoclasts. Methods A tail-suspension (TS) mouse model was used in this study to mimic muscular disuse. ELISA and immunohistochemistry were performed to detect bone and serum LIF levels. Micro-computed tomography (CT) of the mouse femurs was performed to measure three-dimensional bone structure parameters. Fluid shear stress (FSS) and microgravity simulation experiments were performed to study mechanical stress-induced LIF secretion and its resultant effects. Bone marrow macrophages (BMMs) and bone mesenchymal stem cells (BMSCs) were cultured to induce in vitro osteoclastogenesis and osteogenesis, respectively. Results Micro-CT results showed that TS mice exhibited deteriorated bone microstructure and lower serum LIF expression. LIF secretion by osteocytes was promoted by FSS and was repressed in a microgravity environment. Further experiments showed that LIF could elevate the tartrate-resistant acid phosphatase activity in BMM-derived osteoclasts through the STAT3 signaling pathway. LIF also enhanced alkaline phosphatase staining and osteogenesis-related gene expression during the osteogenic differentiation of BMSCs. Conclusion Mechanical loading affected LIF expression levels in osteocytes, thereby altering the balance between osteoclastogenesis and osteogenesis.
Collapse
Affiliation(s)
- Jingke Du
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiancheng Yang
- Department of Spinal Surgery, People's Hospital of Longhua Shenzhen, Shenzhen, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, China
| | - Zihao He
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Arthritis Clinic and Research Center, Peking University People's Hospital, Peking University, Beijing, China
| | - Junqi Cui
- Department of Pathology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqi Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingming Xu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ning Zhao
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengning Yan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanjun Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Williams KM, Leser JM, Gould NR, Joca HC, Lyons JS, Khairallah RJ, Ward CW, Stains JP. TRPV4 calcium influx controls sclerostin protein loss independent of purinergic calcium oscillations. Bone 2020; 136:115356. [PMID: 32272228 PMCID: PMC7605285 DOI: 10.1016/j.bone.2020.115356] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 02/09/2023]
Abstract
Skeletal remodeling is driven in part by the osteocyte's ability to respond to its mechanical environment by regulating the abundance of sclerostin, a negative regulator of bone mass. We have recently shown that the osteocyte responds to fluid shear stress via the microtubule network-dependent activation of NADPH oxidase 2 (NOX2)-generated reactive oxygen species and subsequent opening of TRPV4 cation channels, leading to calcium influx, activation of CaMKII, and rapid sclerostin protein downregulation. In addition to the initial calcium influx, purinergic receptor signaling and calcium oscillations occur in response to mechanical load and prior to rapid sclerostin protein loss. However, the independent contributions of TRPV4-mediated calcium influx and purinergic calcium oscillations to the rapid sclerostin protein downregulation remain unclear. Here, we showed that NOX2 and TRPV4-dependent calcium influx is required for calcium oscillations, and that TRPV4 activation is both necessary and sufficient for sclerostin degradation. In contrast, calcium oscillations are neither necessary nor sufficient to acutely decrease sclerostin protein abundance. However, blocking oscillations with apyrase prevented fluid shear stress induced changes in osterix (Sp7), osteoprotegerin (Tnfrsf11b), and sclerostin (Sost) gene expression. In total, these data provide key mechanistic insights into the way bone cells translate mechanical cues to target a key effector of bone formation, sclerostin.
Collapse
Affiliation(s)
- Katrina M Williams
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jenna M Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicole R Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Humberto C Joca
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - James S Lyons
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Christopher W Ward
- Department of Orthopaedics, University of Maryland School of Nursing, Baltimore, MD 21201, USA.
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
29
|
Carluccio M, Ziberi S, Zuccarini M, Giuliani P, Caciagli F, Di Iorio P, Ciccarelli R. Adult mesenchymal stem cells: is there a role for purine receptors in their osteogenic differentiation? Purinergic Signal 2020; 16:263-287. [PMID: 32500422 DOI: 10.1007/s11302-020-09703-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
The role played by mesenchymal stem cells (MSCs) in contributing to adult tissue homeostasis and damage repair thanks to their differentiation capabilities has raised a great interest, mainly in bone regenerative medicine. The growth/function of these undifferentiated cells of mesodermal origin, located in specialized structures (niches) of differentiated organs is influenced by substances present in this microenvironment. Among them, ancestral and ubiquitous molecules such as adenine-based purines, i.e., ATP and adenosine, may be included. Notably, extracellular purine concentrations greatly increase during tissue injury; thus, MSCs are exposed to effects mediated by these agents interacting with their own receptors when they act/migrate in vivo or are transplanted into a damaged tissue. Here, we reported that ATP modulates MSC osteogenic differentiation via different P2Y and P2X receptors, but data are often inconclusive/contradictory so that the ATP receptor importance for MSC physiology/differentiation into osteoblasts is yet undetermined. An exception is represented by P2X7 receptors, whose expression was shown at various differentiation stages of bone cells resulting essential for differentiation/survival of both osteoclasts and osteoblasts. As well, adenosine, usually derived from extracellular ATP metabolism, can promote osteogenesis, likely via A2B receptors, even though findings from human MSCs should be implemented and confirmed in preclinical models. Therefore, although many data have revealed possible effects caused by extracellular purines in bone healing/remodeling, further studies, hopefully performed in in vivo models, are necessary to identify defined roles for these compounds in favoring/increasing the pro-osteogenic properties of MSCs and thereby their usefulness in bone regenerative medicine.
Collapse
Affiliation(s)
- Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy.,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy
| | - Sihana Ziberi
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy.,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Francesco Caciagli
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100, Chieti, Italy. .,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi, 66100, Chieti, Italy. .,StemTeCh Group, Via L. Polacchi, 66100, Chieti, Italy.
| |
Collapse
|
30
|
Schwebach CL, Kudryashova E, Zheng W, Orchard M, Smith H, Runyan LA, Egelman EH, Kudryashov DS. Osteogenesis imperfecta mutations in plastin 3 lead to impaired calcium regulation of actin bundling. Bone Res 2020; 8:21. [PMID: 32509377 PMCID: PMC7244493 DOI: 10.1038/s41413-020-0095-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/06/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022] Open
Abstract
Mutations in actin-bundling protein plastin 3 (PLS3) emerged as a cause of congenital osteoporosis, but neither the role of PLS3 in bone development nor the mechanisms underlying PLS3-dependent osteoporosis are understood. Of the over 20 identified osteoporosis-linked PLS3 mutations, we investigated all five that are expected to produce full-length protein. One of the mutations distorted an actin-binding loop in the second actin-binding domain of PLS3 and abolished F-actin bundling as revealed by cryo-EM reconstruction and protein interaction assays. Surprisingly, the remaining four mutants fully retained F-actin bundling ability. However, they displayed defects in Ca2+ sensitivity: two of the mutants lost the ability to be inhibited by Ca2+, while the other two became hypersensitive to Ca2+. Each group of the mutants with similar biochemical properties showed highly characteristic cellular behavior. Wild-type PLS3 was distributed between lamellipodia and focal adhesions. In striking contrast, the Ca2+-hyposensitive mutants were not found at the leading edge but localized exclusively at focal adhesions/stress fibers, which displayed reinforced morphology. Consistently, the Ca2+-hypersensitive PLS3 mutants were restricted to lamellipodia, while chelation of Ca2+ caused their redistribution to focal adhesions. Finally, the bundling-deficient mutant failed to co-localize with any F-actin structures in cells despite a preserved F-actin binding through a non-mutation-bearing actin-binding domain. Our findings revealed that severe osteoporosis can be caused by a mutational disruption of the Ca2+-controlled PLS3's cycling between adhesion complexes and the leading edge. Integration of the structural, biochemical, and cell biology insights enabled us to propose a molecular mechanism of plastin activity regulation by Ca2+.
Collapse
Affiliation(s)
- Christopher L. Schwebach
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
- Molecular Cellular and Developmental Biology graduate program, The Ohio State University, Columbus, OH 43210 USA
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Weili Zheng
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908 USA
| | - Matthew Orchard
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Harper Smith
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
- Biophysics graduate program, The Ohio State University, Columbus, OH 43210 USA
| | - Lucas A. Runyan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
| | - Edward H. Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908 USA
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210 USA
- Molecular Cellular and Developmental Biology graduate program, The Ohio State University, Columbus, OH 43210 USA
- Biophysics graduate program, The Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
31
|
Delgado-Ruiz RA, Calvo-Guirado JL, Romanos GE. Effects of occlusal forces on the peri-implant-bone interface stability. Periodontol 2000 2019; 81:179-193. [PMID: 31407438 DOI: 10.1111/prd.12291] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The occlusal forces and their influence on the initiation of peri-implant bone loss or their relationship with peri-implantitis have created discussion during the past 30 years given the discrepancies observed in clinical, animal, and finite element analysis studies. Beyond these contradictions, in the case of an osseointegrated implant, the occlusal forces can influence the implant-bone interface and the cells responsible for the bone remodeling in different ways that may result in the maintenance or loss of the osseointegration. This comprehensive review focuses on the information available about the forces transmitted through the implant-crown system to the implant-bone interface and the mechano-transduction phenomena responsible for the bone cells' behavior and their interactions. Knowledge of the basic molecular biology of the peri-implant bone would help clinicians to understand the complex phenomenon of occlusal forces and their effects on the implant-bone interface, and would allow better control of the negative effects of mechanical stresses, leading to therapy with fewer risks and complications.
Collapse
Affiliation(s)
- Rafael Arcesio Delgado-Ruiz
- Department of Prosthodontics and Digital Technology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Jose Luis Calvo-Guirado
- International Dentistry Research Cathedra, Faculty of Medicine and Dentistry, Universidad Catolica San Antonio De Murcia (UCAM), Murcia, Spain
| | - Georgios E Romanos
- Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA.,Department of Oral Surgery and Implant Dentistry, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|
32
|
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: 59] [Impact Index Per Article: 9.8] [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
|
33
|
Abstract
Signal transducer and activator of transcription 3 (Stat3) is a member of the Stat family of proteins involved in signaling in many different cell types, including osteocytes. Osteocytes are considered major mechanosensing cells in bone due to their intricate dendritic networks able to sense changes in physical force and to orchestrate the response of osteoclasts and osteoblasts. We examined the role of Stat3 in osteocytes by generating mice lacking Stat3 in these cells using the Dmp-1(8kb)-Cre promoter (Stat3cKO mice). Compared to age-matched littermate controls, Stat3cKO mice of either sex (18 weeks old) exhibit reduced bone formation indices, decreased osteoblasts and increased osteoclasts, and altered material properties, without detectable changes in bone mineral density (BMD) or content of either trabecular or cortical bone. In addition, Stat3cKO mice of either sex show significantly decreased load-induced bone formation. Furthermore, pharmacologic inhibition of Stat3 in osteocytes in vitro with WP1066 blocked the increase in cytosolic calcium induced by ATP, a mediator of the cellular responses to sheer stress. WP1066 also increased reactive oxygen species (ROS) production in cultured MLO-Y4 osteocytes. These data demonstrate that Stat3 is a critical mediator of mechanical signals received by osteocytes and suggest that osteocytic Stat3 is a potential therapeutic target to stimulate bone anabolism.
Collapse
|
34
|
Mikolajewicz N, Sehayek S, Wiseman PW, Komarova SV. Transmission of Mechanical Information by Purinergic Signaling. Biophys J 2019; 116:2009-2022. [PMID: 31053261 DOI: 10.1016/j.bpj.2019.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/26/2019] [Accepted: 04/08/2019] [Indexed: 12/27/2022] Open
Abstract
The skeleton constantly interacts and adapts to the physical world. We have previously reported that physiologically relevant mechanical forces lead to small repairable membrane injuries in bone-forming osteoblasts, resulting in release of ATP and stimulation of purinergic (P2) calcium responses in neighboring cells. The goal of this study was to develop a theoretical model describing injury-related ATP and ADP release, their extracellular diffusion and degradation, and purinergic responses in neighboring cells. After validation using experimental data for intracellular free calcium elevations, ATP, and vesicular release after mechanical stimulation of a single osteoblast, the model was scaled to a tissue-level injury to investigate how purinergic signaling communicates information about injuries with varying geometries. We found that total ATP released, peak extracellular ATP concentration, and the ADP-mediated signaling component contributed complementary information regarding the mechanical stimulation event. The total amount of ATP released governed spatial factors, such as the maximal distance from the injury at which purinergic responses were stimulated. The peak ATP concentration reflected the severity of an individual cell injury, allowing to discriminate between minor and severe injuries that released similar amounts of ATP because of differences in injury repair, and determined temporal aspects of the response, such as signal propagation velocity. ADP-mediated signaling became relevant only in larger tissue-level injuries, conveying information about the distance to the injury site and its geometry. Thus, we identified specific features of extracellular ATP and ADP spatiotemporal signals that depend on tissue mechanoresilience and encode the severity, scope, and proximity of the mechanical stimulus.
Collapse
Affiliation(s)
- Nicholas Mikolajewicz
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| | | | - Paul W Wiseman
- Department of Physics, Montreal, Quebec, Canada; Department of Chemistry, McGill University, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children-Canada, Montreal, Quebec, Canada.
| |
Collapse
|
35
|
Kilic O, Yoon A, Shah SR, Yong HM, Ruiz-Valls A, Chang H, Panettieri RA, Liggett SB, Quiñones-Hinojosa A, An SS, Levchenko A. A microphysiological model of the bronchial airways reveals the interplay of mechanical and biochemical signals in bronchospasm. Nat Biomed Eng 2019; 3:532-544. [PMID: 31150010 PMCID: PMC6653686 DOI: 10.1038/s41551-019-0366-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 02/07/2019] [Indexed: 01/08/2023]
Abstract
In asthma, airway smooth muscle (ASM) contraction and the subsequent decrease in airflow involve a poorly understood set of mechanical and biochemical events. Organ-level and molecular-scale models of the airway are frequently based on purely mechanical or biochemical considerations and do not account for physiological mechanochemical couplings. Here, we present a microphysiological model of the airway that allows for the quantitative analysis of the interactions between mechanical and biochemical signals triggered by compressive stress on epithelial cells. We show that a mechanical stimulus mimicking a bronchospastic challenge triggers the marked contraction and delayed relaxation of ASM, and that this is mediated by the discordant expression of cyclooxygenase genes in epithelial cells and regulated by the mechanosensor and transcriptional co-activator YAP (Yes-associated protein). A mathematical model of the intercellular feedback interactions recapitulates aspects of obstructive disease of the airways, including pathognomonic features of severe, difficult-to-treat asthma. The microphysiological model could be used to investigate the mechanisms of asthma pathogenesis and to develop therapeutic strategies that disrupt the positive feedback loop that leads to persistent airway constriction.
Collapse
Affiliation(s)
- Onur Kilic
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Arum Yoon
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sagar R Shah
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hwan Mee Yong
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alejandro Ruiz-Valls
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hao Chang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Reynold A Panettieri
- Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Stephen B Liggett
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA.,Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | - Steven S An
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA. .,Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA. .,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea.
| | - Andre Levchenko
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA. .,Department of Biomedical Engineering, Yale University, New Haven, CT, USA. .,Yale Systems Biology Institute, Yale University, West Haven, CT, USA.
| |
Collapse
|
36
|
Villaseñor A, Aedo-Martín D, Obeso D, Erjavec I, Rodríguez-Coira J, Buendía I, Ardura JA, Barbas C, Gortazar AR. Metabolomics reveals citric acid secretion in mechanically-stimulated osteocytes is inhibited by high glucose. Sci Rep 2019; 9:2295. [PMID: 30783155 PMCID: PMC6381120 DOI: 10.1038/s41598-018-38154-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/19/2018] [Indexed: 01/15/2023] Open
Abstract
Osteocytes are the main cells of bone tissue and play a crucial role in bone formation and resorption. Recent studies have indicated that Diabetes Mellitus (DM) affects bone mass and potentially causes higher bone fracture risk. Previous work on osteocyte cell cultures has demonstrated that mechanotransduction is impaired after culture under diabetic pre-conditioning with high glucose (HG), specifically osteoclast recruitment and differentiation. The aim of this study was to analyze the extracellular metabolic changes of osteocytes regarding two conditions: pre-conditioning to either basal levels of glucose (B), mannitol (M) or HG cell media, and mechanical stimulation by fluid flow (FF) in contrast to static condition (SC). Secretomes were analyzed using Liquid Chromatography and Capillary Electrophoresis both coupled to Mass Spectrometry (LC-MS and CE-MS, respectively). Results showed the osteocyte profile was very similar under SC, regardless of their pre-conditioning treatment, while, after FF stimulation, secretomes followed different metabolic signatures depending on the pre-conditioning treatment. An important increment of citrate pointed out that osteocytes release citrate outside of the cell to induce osteoblast activation, while HG environment impaired FF effect. This study demonstrates for the first time that osteocytes increase citrate excretion under mechanical stimulation, and that HG environment impaired this effect.
Collapse
Affiliation(s)
- Alma Villaseñor
- IMMA, Institute of Applied Molecular Medicine, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Daniel Aedo-Martín
- IMMA, Institute of Applied Molecular Medicine, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain
| | - David Obeso
- IMMA, Institute of Applied Molecular Medicine, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Igor Erjavec
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Juan Rodríguez-Coira
- IMMA, Institute of Applied Molecular Medicine, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Irene Buendía
- IMMA, Institute of Applied Molecular Medicine, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Juan Antonio Ardura
- IMMA, Institute of Applied Molecular Medicine, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.,Basic Medical Sciences Department, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Arancha R Gortazar
- IMMA, Institute of Applied Molecular Medicine, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain. .,Basic Medical Sciences Department, School of Medicine, CEU San Pablo University, Campus Monteprincipe, Boadilla del Monte, 28668, Madrid, Spain.
| |
Collapse
|
37
|
Ma Q, Ma Z, Liang M, Luo F, Xu J, Dou C, Dong S. The role of physical forces in osteoclastogenesis. J Cell Physiol 2019; 234:12498-12507. [PMID: 30623443 DOI: 10.1002/jcp.28108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022]
Abstract
The movements of life at every level from organs, tissues, cells to sub-cells, are all conducted in certain physical environments. In the human body, skeletal tissue among all connective tissues is influenced the most by physical forces. Studying the biological behavior of bone cells under different physical environments is helpful in further understanding bone homeostasis and metabolism. Among all bone cells, osteoclast (OC) and OC steered bone remodeling is one of the key points in bone metabolism. In the past few decades, people's understanding of OC was mostly limited to its involvement of bone resorption under physiological and pathological conditions. However, more and more studies started to focus on how physical forces affect the formation and differentiation of OC. This review tries to illustrate the knowledge up to date about how osteoclastogenesis is regulated by physical forces through direct and indirect ways, including fluid shear force, compressive force, and microgravity. The direct way describes the straightforward effects produced by different forces in osteoclastogenesis, whereas the indirect way describes the effects of different forces in osteoclastogenesis through regulation of other bone cells when a certain force is applied. Molecular mechanisms were analyzed and reviewed in both direct and indirect regulation by different forces. Finally, we discussed the status quo and tendency of related research, as well as other unresolved issues, and some future prospects.
Collapse
Affiliation(s)
- Qinyu Ma
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| | - Zaisong Ma
- Department of Orthopedics, General Hospital of Xinjiang Command, Urumqi, Xinjiang, China
| | - Mengmeng Liang
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| |
Collapse
|
38
|
Hinton PV, Rackard SM, Kennedy OD. In Vivo Osteocyte Mechanotransduction: Recent Developments and Future Directions. Curr Osteoporos Rep 2018; 16:746-753. [PMID: 30406580 DOI: 10.1007/s11914-018-0485-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Mechanical loading is an essential stimulus for skeletal tissues. Osteocytes are primarily responsible for sensing mechanical stimuli in bone and for orchestrating subsequent responses. This is critical for maintaining homeostasis, and responding to injury/disease. The osteocyte mechanotransduction pathway, and the downstream effects it mediates, is highly complex. In vivo models have proved invaluable in understanding this process. This review summarizes the commonly used models, as well as more recently developed ones, and describes how they are used to address emerging questions in the field. RECENT FINDINGS Minimally invasive animal models can be used to determine mechanisms of osteocyte mechanotransduction, at the cell and molecular level, while simultaneously reducing potentially confounding responses such as inflammation/wound-healing. The details of osteocyte mechanotransduction in bone are gradually becoming clearer. In vivo model systems are a key tool in pursing this question. Advances in this field are explored and discussed in this review.
Collapse
Affiliation(s)
- Paige V Hinton
- Department of Anatomy & Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland
| | - Susan M Rackard
- School of Veterinary Medicine, Veterinary Science Centre, University College Dublin, Dublin 4, Ireland
| | - Oran D Kennedy
- Department of Anatomy & Tissue Engineering Research Group, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
39
|
Mikolajewicz N, Zimmermann EA, Willie BM, Komarova SV. Mechanically stimulated ATP release from murine bone cells is regulated by a balance of injury and repair. eLife 2018; 7:37812. [PMID: 30324907 PMCID: PMC6205812 DOI: 10.7554/elife.37812] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/28/2018] [Indexed: 02/06/2023] Open
Abstract
Bone cells sense and actively adapt to physical perturbations to prevent critical damage. ATP release is among the earliest cellular responses to mechanical stimulation. Mechanical stimulation of a single murine osteoblast led to the release of 70 ± 24 amole ATP, which stimulated calcium responses in neighboring cells. Osteoblasts contained ATP-rich vesicles that were released upon mechanical stimulation. Surprisingly, interventions that promoted vesicular release reduced ATP release, while inhibitors of vesicular release potentiated ATP release. Searching for an alternative ATP release route, we found that mechanical stresses induced reversible cell membrane injury in vitro and in vivo. Ca2+/PLC/PKC-dependent vesicular exocytosis facilitated membrane repair, thereby minimizing cell injury and reducing ATP release. Priming cellular repair machinery prior to mechanical stimulation reduced subsequent membrane injury and ATP release, linking cellular mechanosensitivity to prior mechanical exposure. Thus, our findings position ATP release as an integrated readout of membrane injury and repair. Athletes' skeletons get stronger with training, while bones weaken in people who cannot move or in astronauts experiencing weightlessness. This is because bone cells thrive when exposed to forces. When a bone cell is exposed to a physical force, the first thing that happens is the release of the energy-rich molecule called ATP into the space outside the cell. This molecule then binds to the neighboring cell to unleash a cascade of responses. ATP can exit the cell either through special canals in the cell membrane or released in tiny pod-like structures called vesicles. It is known that strong forces can injure the cell membrane and cause ATP to spill out. However, it is less clear how ATP is released when cells are subjected to regular forces. Mikolajewicz et al. investigated whether ATP exits through injured membranes of cells experiencing regular forces. Bone cells grown in the laboratory were gently poked with a glass needle or placed in a turbulent fluid to simulate forces experienced in the body. Dyes and fluorescent imaging techniques were used to observe the movement of vesicles and calculate the concentration of ATP in these cells. The experiments showed that regular forces in the body do indeed injure the cell membranes and cause ATP to spill out. But importantly, the cells repaired the injuries quickly by releasing vesicles that patch the wound. As soon as the membrane is sealed, ATP stops coming out. From the first injury, cells adapted and quickly strengthened their membrane and repair system to be more resilient against future forces. This process was also seen in the shin bones of mice. These results are important because knowing how bone cells sense, respond and convert physical forces can help us develop treatments for astronauts, the injured and aged.
Collapse
Affiliation(s)
- Nicholas Mikolajewicz
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| | - Elizabeth A Zimmermann
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Department of Pediatric Surgery, Montreal, Quebec, Canada
| | - Bettina M Willie
- Shriners Hospital for Children - Canada, Montreal, Quebec, Canada.,Department of Pediatric Surgery, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospital for Children - Canada, Montreal, Quebec, Canada
| |
Collapse
|
40
|
Laiuppa JA, Santillán GE. Involvement of GSK3/β-catenin in the action of extracellular ATP on differentiation of primary cultures from rat calvaria into osteoblasts. J Cell Biochem 2018; 119:8378-8388. [PMID: 29932242 DOI: 10.1002/jcb.27037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/05/2018] [Indexed: 11/08/2022]
Abstract
Modulation of purinergic receptors play an important role in the regulation of osteoblasts differentiation and bone formation. In this study, we investigated the involvement of the GSK3/βcatenin signaling in the action of ATPγ-S on osteogenic differentiation of primary cell cultures from rat calvaria. Our results indicate that the cell treatment with 10 or 100 µM ATPγ-S for 96 h increase the cytoplasmic levels of β-catenin and its translocation to nucleus respect to control. A similar effect was observed after cell treatment with the GSK3 inhibitor LiCl (10 mM). Cell treatments with 4-10 mM LiCl significantly stimulated ALP activity respect to control at 4 and 7 days, suggesting that inhibition of GSK-3 mediates osteoblastic differentiation of rat calvarial cells. Effects comparison between ATP and LiCl shown that ALP activity was significantly increased by 10 µM ATPγ-S and decreased by 10 mM LiCl at 10 day of treatment, respect to control, suggesting that the effect of ATPγ-S was less potent but more persistent than of LiCl in stimulating this osteogenic marker in calvarial cells. Cell culture mineralization was significantly increased by treatment with 10 µM ATPγ-S and decreased by 10 mM LiCl, respect to control. In together, these results suggest that GSK3 inhibition is involved in ATPγ-S action on rat calvarial cell differentiation into osteoblasts at early steadies. In addition such inhibition by LiCl appear promote osteoblasts differentiation at beginning but has a deleterious effect on its function at later steadies as the extracellular matrix mineralization.
Collapse
Affiliation(s)
- Juan A Laiuppa
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, INBIOSUR-CONICET, Bahía Blanca, Argentina
| | - Graciela E Santillán
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, INBIOSUR-CONICET, Bahía Blanca, Argentina
| |
Collapse
|
41
|
Tassinary JAF, Lunardelli A, Basso BDS, Dias HB, Catarina AV, Stülp S, Haute GV, Martha BA, Melo DADS, Nunes FB, Donadio MVF, Oliveira JRD. Low-intensity pulsed ultrasound (LIPUS) stimulates mineralization of MC3T3-E1 cells through calcium and phosphate uptake. ULTRASONICS 2018; 84:290-295. [PMID: 29182945 DOI: 10.1016/j.ultras.2017.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 06/07/2023]
Abstract
The present study aimed to evaluate the effect of low-intensity pulsed ultrasound (LIPUS) on pre-osteoblast mineralization using in vitro bioassays. Pre-osteoblastic MC3T3-E1 cells were exposed to LIPUS at 1 MHz frequency, 0.2 W/cm2 intensity and 20% duty cycle for 30 min. The analyses were carried out up to 336 h (14 days) after exposure. The concentration of collagen, phosphate, alkaline phosphatase, calcium and transforming growth factor beta 1 (TGF-β1) in cell supernatant and the presence of calcium deposits in the cells were analyzed. Our results showed that LIPUS promotes mineralized nodules formation. Collagen, phosphate, and calcium levels were decreased in cell supernatant at 192 h after LIPUS exposure. However, alkaline phosphatase and TGF-β1 concentrations remained unchanged. Therapeutic pulsed ultrasound is capable of stimulating differentiation and mineralization of pre-osteoblastic MC3T3-E1 cells by calcium and phosphate uptake with consequent hydroxyapatite formation.
Collapse
Affiliation(s)
- João Alberto Fioravante Tassinary
- Univates, Lajeado, Rio Grande do Sul, Brazil; Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Adroaldo Lunardelli
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil; Centro Universitário Ritter dos Reis (UniRitter), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruno de Souza Basso
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Henrique Bregolin Dias
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Anderson Velasque Catarina
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Gabriela Viegas Haute
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bianca Andrade Martha
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Denizar Alberto da Silva Melo
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Bordignon Nunes
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
42
|
Understanding the functional role of genistein in the bone differentiation in mouse osteoblastic cell line MC3T3-E1 by RNA-seq analysis. Sci Rep 2018; 8:3257. [PMID: 29459627 PMCID: PMC5818530 DOI: 10.1038/s41598-018-21601-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 02/07/2018] [Indexed: 01/23/2023] Open
Abstract
Genistein, a phyto-estrogen, can potentially replace endogenous estrogens in postmenopausal women, but the underlying molecular mechanisms remain incompletely understood. To obtain insight into the effect of genistein on bone differentiation, RNA sequencing (RNA-seq) analysis was used to detect differentially expressed genes (DEGs) in genistein-treated vs. untreated MC3T3-E1 mouse osteoblastic cells. Osteoblastic cell differentiation was monitored by measuring osteoblast differentiation factors (ALP production, bone mineralization, and expression of osteoblast differentiation markers). From RNA-seq analysis, a total of 132 DEGs (including 52 up-regulated and 80 down-regulated genes) were identified in genistein-treated cells (FDR q-value < 0.05 and fold change > 1.5). KEGG pathway and Gene Ontology (GO) enrichment analyses were performed to estimate the biological functions of DEGs and demonstrated that these DEGs were highly enriched in functions related to chemotactic cytokines. The functional relevance of DEGs to genistein-induced osteoblastic cell differentiation was further evaluated by siRNA-mediated knockdown in MC3T3-E1 cells. These siRNA knockdown experiments (of the DEGs validated by real-time qPCR) demonstrated that two up-regulated genes (Ereg and Efcab2) enhance osteoblastic cell differentiation, while three down-regulated genes (Hrc, Gli, and Ifitm5) suppress the differentiation. These results imply their major functional roles in bone differentiation regulated by genistein.
Collapse
|
43
|
Sindhavajiva PR, Sastravaha P, Arksornnukit M, Pavasant P. Intermittent compressive force induces human mandibular-derived osteoblast differentiation via WNT/β-catenin signaling. J Cell Biochem 2018; 119:3474-3485. [PMID: 29143994 DOI: 10.1002/jcb.26519] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/13/2017] [Indexed: 02/02/2023]
Abstract
Mechanical force induces an efflux of ATP that regulates osteoblast differentiation. However, the effect of mechanical force-induced ATP efflux on WNT/β-catenin signaling remains unclarified. The aim of this study was to investigate the effect of intermittent compressive force (ICF) and ICF-induced extracellular ATP on osteoblast differentiation via WNT/β-catenin signaling in human mandibular-derived osteoblast precursors (hMOBPs). The hMOBPs were subjected to ICF (1.5 g/cm2 , 0.3 Hz) for 20 h. To investigate the role of ATP, Apyrase (0.5 units/mL), an enzyme that hydrolyzes ATP, was added 30 min before ICF was applied. The extracellular ATP levels were measured immediately after ICF was removed. The mRNA expression of osteogenic related genes, including WNT was evaluated via quantitative real time polymerase chain reaction. In vitro mineralization was determined by Alizarin Red S staining. The localization of β-catenin was detected using immunofluorescence staining and lentiviral-TOP-dGFP reporter assay. The results demonstrated that ICF increased ATP efflux and in vitro mineralization by hMOBPs. In addition, OSX, ALP, and WNT3A mRNA expression and β-catenin nuclear translocation increased when ICF was applied. The upregulation of these genes was reduced by Apyrase, suggesting the role of ICF-induced ATP on osteoblast differentiation. Notably, ICF altered the mRNA expression of purinergic 2X receptors (P2XRs). A P2X1R antagonist (NF449) downregulated ICF-induced WNT3A, OSX, and ALP mRNA expression. Moreover, when 25 μM α, β-meATP, a P2X1R agonist, was added, WNT3A, and OSX expression increased. In conclusion, our results demonstrate that ICF-induced ATP enhanced hMOBP differentiation. This enhancement was associated with WNT/β-catenin signaling and P2X1R activation.
Collapse
Affiliation(s)
- Pimrumpai R Sindhavajiva
- Graduate Program in Prosthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Panunn Sastravaha
- Department of Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Mansuang Arksornnukit
- Department of Prosthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
44
|
Steward AJ, Kelly DJ, Wagner DR. Purinergic Signaling Regulates the Transforming Growth Factor-β3-Induced Chondrogenic Response of Mesenchymal Stem Cells to Hydrostatic Pressure. Tissue Eng Part A 2017; 22:831-9. [PMID: 27137792 DOI: 10.1089/ten.tea.2015.0047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Although hydrostatic pressure (HP) is known to regulate chondrogenic differentiation of mesenchymal stromal/stem cells (MSCs), improved insight into the mechanotransduction of HP may form the basis for novel tissue engineering strategies. Previously, we demonstrated that matrix stiffness and calcium ion (Ca(++)) mobility regulate the mechanotransduction of HP; however, the mechanisms, by which these Ca(++) signaling pathways are initiated, are currently unknown. The purinergic pathway, in which adenosine triphosphate (ATP) is released and activates P-receptors to initiate Ca(++) signaling, plays a key role in the mechanotransduction of compression, but has yet to be investigated with regard to HP. Therefore, the objective of this study was to investigate the interplay between purinergic signaling, matrix stiffness, and the chondrogenic response of MSCs to HP. Porcine bone marrow-derived MSCs were seeded into soft or stiff agarose hydrogels and subjected to HP (10 MPa at 1 Hz for 4 h/d for 21 days) or kept in free swelling conditions. Stiff constructs were incubated with pharmacological inhibitors of extracellular ATP, P2 receptors, or hemichannels, or without any inhibitors as a control. As with other loading modalities, HP significantly increased ATP release in the control group; however, inhibition of hemichannels completely abrogated this response. The increase in sulfated glycosaminoglycan (sGAG) synthesis and vimentin reorganization observed in the control group in response to HP was suppressed in the presence of all three inhibitors, suggesting that purinergic signaling is involved in the mechanoresponse of MSCs to HP. Interestingly, ATP was released from both soft and stiff hydrogels in response to HP, but HP only enhanced chondrogenesis in the stiff hydrogels, indicating that matrix stiffness may act downstream of purinergic signaling to regulate the mechanoresponse of MSCs to HP. Addition of exogenous ATP did not replicate the effects of HP on chondrogenesis, suggesting that mechanisms other than purinergic signaling also regulate the response of MSCs to HP.
Collapse
Affiliation(s)
- Andrew J Steward
- 1 Bioengineering Graduate Program, Department of Aerospace and Mechanical Engineering, University of Notre Dame , Notre Dame, Indiana.,2 Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute , Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- 2 Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute , Trinity College Dublin, Dublin, Ireland .,3 Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin , Dublin, Ireland .,4 Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin , Dublin, Ireland
| | - Diane R Wagner
- 5 Department of Mechanical Engineering, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana.,6 Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| |
Collapse
|
45
|
PYK2 mediates BzATP-induced extracellular matrix proteins synthesis. Biochem Biophys Res Commun 2017; 494:663-667. [PMID: 29061307 DOI: 10.1016/j.bbrc.2017.10.107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 10/20/2017] [Indexed: 10/25/2022]
Abstract
Mechanical stimuli such as fluid shear and cyclic tension force induced extracellular adenosine triphosphate (ATP) release in osteoblasts. In particular, cyclic tension force-induced ATP enhances bone formation through P2X7 activation. Proline-rich tyrosine kinase 2 (PYK2) mediate osteoblasts differentiation is induced by mechanical stimuli. Furthermore, activation of PYK2 also was a response to integrin by mechanical stimuli. Extracellular matrix protein (ECMP)s, which are important factors for bone formation are expressed by osteoblasts. However, the effect of the interaction of 2'(3)-Ο-(4-Benzoylbenzoyl) adenosine-5'-triphosphate (BzATP), which is the agonist of the mechanosensitive receptor P2X7, with PYK2 on ECMP production is poorly understood. Thus, our purpose was to investigate the effects of PYK2 on BzATP-induced ECMP production in osteoblasts. BzATP increased phospho-PYK2 protein expression on days 3 and 7 of culture. Furthermore, the PYK2 inhibitor PF431394 inhibited the stimulatory effect of BzATP on the expression of type I collagen, bone sialoprotein and osteocalcin expression. PF431396 did not inhibit the stimulatory effect of BzATP on osteopontin (OPN) mRNA expression. These results suggest that mechanical stimuli activate P2X7 might induce ECMPs expression through PYK2 except in the case of OPN expression. Altogether, mechanical stimuli-induced ECMPs production might be implicated by extracellular ATP secretion or integrin via PYK2 activation.
Collapse
|
46
|
Heo SJ, Han WM, Szczesny SE, Cosgrove BD, Elliott DM, Lee DA, Duncan RL, Mauck RL. Mechanically Induced Chromatin Condensation Requires Cellular Contractility in Mesenchymal Stem Cells. Biophys J 2017; 111:864-874. [PMID: 27558729 DOI: 10.1016/j.bpj.2016.07.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/27/2016] [Accepted: 07/11/2016] [Indexed: 02/07/2023] Open
Abstract
Mechanical cues play important roles in directing the lineage commitment of mesenchymal stem cells (MSCs). In this study, we explored the molecular mechanisms by which dynamic tensile loading (DL) regulates chromatin organization in this cell type. Our previous findings indicated that the application of DL elicited a rapid increase in chromatin condensation through purinergic signaling mediated by ATP. Here, we show that the rate and degree of condensation depends on the frequency and duration of mechanical loading, and that ATP release requires actomyosin-based cellular contractility. Increases in baseline cellular contractility via the addition of an activator of G-protein coupled receptors (lysophosphatidic acid) induced rapid ATP release, resulting in chromatin condensation independent of loading. Conversely, inhibition of contractility through pretreatment with either a RhoA/Rock inhibitor (Y27632) or MLCK inhibitor (ML7) abrogated ATP release in response to DL, blocking load-induced chromatin condensation. With loading, ATP release occurred very rapidly (within the first 10-20 s), whereas changes in chromatin occurred at a later time point (∼10 min), suggesting a downstream biochemical pathway mediating this process. When cells were pretreated with blockers of the transforming growth factor (TGF) superfamily, purinergic signaling in response to DL was also eliminated. Further analysis showed that this pretreatment decreased contractility, implicating activity in the TGF pathway in the establishment of the baseline contractile state of MSCs (in the absence of exogenous ligands). These data indicate that chromatin condensation in response to DL is regulated through the interplay between purinergic and RhoA/Rock signaling, and that ligandless activity in the TGF/bone morphogenetic proteins signaling pathway contributes to the establishment of baseline contractility in MSCs.
Collapse
Affiliation(s)
- Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Woojin M Han
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Spencer E Szczesny
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, Pennsylvania
| | - Brian D Cosgrove
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, Pennsylvania
| | - Dawn M Elliott
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - David A Lee
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Randall L Duncan
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware; Department of Biological Sciences, University of Delaware, Newark, Delaware
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, Pennsylvania.
| |
Collapse
|
47
|
Cell culture: complications due to mechanical release of ATP and activation of purinoceptors. Cell Tissue Res 2017; 370:1-11. [PMID: 28434079 PMCID: PMC5610203 DOI: 10.1007/s00441-017-2618-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/21/2017] [Indexed: 12/11/2022]
Abstract
There is abundant evidence that ATP (adenosine 5′-triphosphate) is released from a variety of cultured cells in response to mechanical stimulation. The release mechanism involved appears to be a combination of vesicular exocytosis and connexin and pannexin hemichannels. Purinergic receptors on cultured cells mediate both short-term purinergic signalling of secretion and long-term (trophic) signalling such as proliferation, migration, differentiation and apoptosis. We aim in this review to bring to the attention of non-purinergic researchers using tissue culture that the release of ATP in response to mechanical stress evoked by the unavoidable movement of the cells acting on functional purinergic receptors on the culture cells is likely to complicate the interpretation of their data.
Collapse
|
48
|
Nagao M, Tanabe N, Manaka S, Naito M, Sekino J, Takayama T, Kawato T, Torigoe G, Kato S, Tsukune N, Maeno M, Suzuki N, Sato S. LIPUS suppressed LPS-induced IL-1α through the inhibition of NF-κB nuclear translocation via AT1-PLCβ pathway in MC3T3-E1 cells. J Cell Physiol 2017; 232:3337-3346. [PMID: 28063227 DOI: 10.1002/jcp.25777] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 01/05/2017] [Indexed: 12/17/2022]
Abstract
Inflammatory cytokines, interleukin (IL)-1, IL-6, and TNF-α, are involved in inflammatory bone diseases such as rheumatoid osteoarthritis and periodontal disease. Particularly, periodontal disease, which destroys alveolar bone, is stimulated by lipopolysaccharide (LPS). Low-intensity pulsed ultrasound (LIPUS) is used for bone healing in orthopedics and dental treatments. However, the mechanism underlying effects of LIPUS on LPS-induced inflammatory cytokine are not well understood. We therefore aimed to investigate the role of LIPUS on LPS-induced IL-1α production. Mouse calvaria osteoblast-like cells MC3T3-E1 were incubated in the presence or absence of LPS (Porphyromonas gingivalis), and then stimulated with LIPUS for 30 min/day. To investigate the role of LIPUS, we determined the expression of IL-1α stimulated with LIPUS and treated with an angiotensin II receptor type 1 (AT1) antagonist, Losartan. We also investigate to clarify the pathway of LIPUS, we transfected siRNA silencing AT1 (siAT1) in MC3T3-E1. LIPUS inhibited mRNA and protein expression of LPS-induced IL-1α. LIPUS also reduced the nuclear translocation of NF-κB by LPS-induced IL-1α. Losartan and siAT1 blocked all the stimulatory effects of LIPUS on IL-1α production and IL-1α-mediated NF-κB translocation induced by LPS. Furthermore, PLCβ inhibitor U73122 recovered NF-κB translocation. These results suggest that LIPUS inhibits LPS-induced IL-1α via AT1-PLCβ in osteoblasts. We exhibit that these findings are in part of the signaling pathway of LIPUS on the anti-inflammatory effects of IL-1α expression.
Collapse
Affiliation(s)
- Mayu Nagao
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Natsuko Tanabe
- Department of Biochemistry, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Soichiro Manaka
- Department of Periodontology, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Masako Naito
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Department of Anatomy, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Jumpei Sekino
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Tadahiro Takayama
- Department of Periodontology, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Functional Morphology, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Takayuki Kawato
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Department of Oral Health Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Go Torigoe
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | | | - Naoya Tsukune
- Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Masao Maeno
- Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Department of Oral Health Sciences, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Naoto Suzuki
- Department of Biochemistry, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| | - Shuichi Sato
- Department of Periodontology, Nihon University Graduate School of Dentistry, Tokyo, Japan.,Division of Functional Morphology, Dental Research Center, Nihon University Graduate School of Dentistry, Tokyo, Japan
| |
Collapse
|
49
|
Xiong L, Jung JU, Guo HH, Pan JX, Sun XD, Mei L, Xiong WC. Osteoblastic Lrp4 promotes osteoclastogenesis by regulating ATP release and adenosine-A 2AR signaling. J Cell Biol 2017; 216:761-778. [PMID: 28193701 PMCID: PMC5350517 DOI: 10.1083/jcb.201608002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/23/2016] [Accepted: 01/10/2017] [Indexed: 02/05/2023] Open
Abstract
Lrp4 is mutated in patients with high-bone-mass diseases. Loss of Lrp4 in osteoblasts (OBs) increases bone formation by OBs and decreases bone resorption by osteoclasts through an unclear mechanism. Xiong et al. show that overproduction of extracellular adenosine in Lrp4-deficient OBs, which are derived from ATP hydrolysis and signals through A2AR and RANK, may underlie Lrp4 regulation of osteoclastogenesis. Bone homeostasis depends on the functional balance of osteoblasts (OBs) and osteoclasts (OCs). Lrp4 is a transmembrane protein that is mutated in patients with high bone mass. Loss of Lrp4 in OB-lineage cells increases bone mass by elevating bone formation by OBs and reducing bone resorption by OCs. However, it is unclear how Lrp4 deficiency in OBs impairs osteoclastogenesis. Here, we provide evidence that loss of Lrp4 in the OB lineage stabilizes the prorenin receptor (PRR) and increases PRR/V-ATPase–driven ATP release, thereby enhancing the production of the ATP derivative adenosine. Both pharmacological and genetic inhibition of adenosine-2A receptor (A2AR) in culture and Lrp4 mutant mice diminishes the osteoclastogenic deficit and reduces trabecular bone mass. Furthermore, elevated adenosine-A2AR signaling reduces receptor activator of nuclear factor κB (RANK)–mediated osteoclastogenesis. Collectively, these results identify a mechanism by which osteoblastic Lrp4 controls osteoclastogenesis, reveal a cross talk between A2AR and RANK signaling in osteoclastogenesis, and uncover an unrecognized pathophysiological mechanism of high-bone-mass disorders.
Collapse
Affiliation(s)
- Lei Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - Ji-Ung Jung
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta, GA 30912
| | - Hao-Han Guo
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - Jin-Xiu Pan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - Xiang-Dong Sun
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, GA 30912.,Department of Neurology, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, GA 30912 .,Department of Neurology, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| | - Wen-Cheng Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta, GA 30912 .,Department of Neurology, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30912
| |
Collapse
|
50
|
Kodama D, Hirai T, Kondo H, Hamamura K, Togari A. Bidirectional communication between sensory neurons and osteoblasts in an in vitro coculture system. FEBS Lett 2017; 591:527-539. [PMID: 28094440 DOI: 10.1002/1873-3468.12561] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 12/30/2016] [Accepted: 01/11/2017] [Indexed: 12/18/2022]
Abstract
Recent studies have revealed that the sensory nervous system is involved in bone metabolism. However, the mechanism of communication between neurons and osteoblasts is yet to be elucidated. In this study, we investigated the signaling pathways between sensory neurons of the dorsal root ganglion (DRG) and the osteoblast-like MC3T3-E1 cells using an in vitro coculture system. Our findings indicate that signal transduction from DRG-derived neurons to MC3T3-E1 cells is suppressed by antagonists of the AMPA receptor and the NK1 receptor. Conversely, signal transduction from MC3T3-E1 cells to DRG-derived neurons is suppressed by a P2X7 receptor antagonist. Our results suggest that these cells communicate with each other by exocytosis of glutamate, substance P in the efferent signal, and ATP in the afferent signal.
Collapse
Affiliation(s)
- Daisuke Kodama
- Laboratory of Neuropharmacology, School of Pharmacy, Aichi-Gakuin University, Chikusa-ku, Nagoya, Japan.,Department of Pharmacology, School of Dentistry, Aichi-Gakuin University, Chikusa-ku, Nagoya, Japan
| | - Takao Hirai
- Laboratory of Medicinal Resources, School of Pharmacy, Aichi-Gakuin University, Chikusa-ku, Nagoya, Japan
| | - Hisataka Kondo
- Department of Pharmacology, School of Dentistry, Aichi-Gakuin University, Chikusa-ku, Nagoya, Japan
| | - Kazunori Hamamura
- Department of Pharmacology, School of Dentistry, Aichi-Gakuin University, Chikusa-ku, Nagoya, Japan
| | - Akifumi Togari
- Department of Pharmacology, School of Dentistry, Aichi-Gakuin University, Chikusa-ku, Nagoya, Japan
| |
Collapse
|