1
|
Metzger W, Ammo T, Sossong D, Bubel M, Mattes C, Stumpf H, Später T, Laschke MW, Pohlemann T. Establishing a simple protocol to induce the osteogenic differentiation of MC3T3-E1 cells in 2D and its transfer to 3D spheroid cultures. Biotech Histochem 2025:1-14. [PMID: 40265253 DOI: 10.1080/10520295.2025.2489501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
The murine cell line MC3T3-E1 is used in many in vitro studies in bone-related research, but different protocols to induce its osteogenic differentiation have been reported. The aim of this study was to identify the best mixture of osteogenic supplements to induce osteogenic differentiation of MC3T3-E1 subclone 4 cells in a two-dimensional cell culture setup. As spheroids as three-dimensional cell aggregates are of increasing importance, we also present a simple method to generate osteogenic differ.entiated spheroids on this basis. Three different mixtures of osteogenic supplements were used to induce osteogenic differentiation for up to 28 days. Osteogenic differentiation was monitored by alizarin red and von Kossa staining, energy dispersive X-ray (EDX) analysis, and real-time quantitative PCR analysis of osteogenic marker genes. Spheroids were generated from osteogenic differentiated cells by liquid overlay technique. The use of 5 mM β-glycerophosphate, 10 nM dexamethasone, and 50 µg/mL ascorbic acid was able to induce osteogenic differentiation of MC3T3-E1 cells within 14 days, as shown by strong positive signals in both staining methods. Scanning electron microscopy revealed extracellular secretions on the membranes of differentiated cells with a significantly increased calcium content of 16.4 ± 2.4% and a phosphorus content of 10.1 ± 1.1%, as shown by energy dispersive X-ray analysis. Differentiated MC3T3-E1 cells could be detached by incubation in AccuMax for 10 min and spheroids were generated from this cell suspension on day 14. Significant upregulation of the osteogenic markers Sp7, osteocalcin, and bone sialoprotein was detected by real-time quantitative PCR analysis of these spheroids. In addition to other reports in the literature describing osteogenic differentiation of spheroids, we were able to show that it is also possible to generate spheroids from osteogenically differentiated two-dimensional cell cultures, which are easier to handle. Thus, there are indeed several ways to generate osteogenic differentiated MC3T3-E1 spheroids.
Collapse
Affiliation(s)
- W Metzger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - T Ammo
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - D Sossong
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - M Bubel
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - C Mattes
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - H Stumpf
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
| | - T Später
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - M W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - T Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
2
|
Izumiya M, Nobuoka H, Endo H, Ueno R, Mimura M, Saito N, Haniu H. Genetic Profiling of MC3T3-E1 Cells in Different Media: Implications for In Vitro Screening Development. Biomedicines 2025; 13:489. [PMID: 40002902 PMCID: PMC11853507 DOI: 10.3390/biomedicines13020489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The translation of in vitro biomaterial evaluations into successful clinical applications often fails due to discrepancies with in vivo results. Previously, we demonstrated that differences in culture medium conditions influence the bone formation process. This study aimed to investigate the influence of culture media on gene expression during calcification induction in osteoblasts. Methods: Using MC3T3-E1 cells cultured in α Minimum Essential Medium without L-ascorbic acid (αMEM(-)) and Dulbecco's Modified Eagle Medium (DMEM), we screened gene expression profiles through microarray analysis and validated key findings with quantitative PCR. Additionally, we compared these gene expression patterns with those in primary osteoblasts (POBs) cultured under the same medium conditions. Results: The results revealed distinct gene expression profiles in MC3T3-E1 cells depending on the culture medium, while POBs exhibited minimal differences between media, except for the gene Alpl. In αMEM(-), Alpl expression in POBs was significantly increased approximately 4-fold via calcification stimulation (p < 0.0001). POBs cultured in DMEM showed calcification appearance differing from the αMEM(-) condition, even though no significant increase in Alpl expression via calcification stimulation was observed. Conclusions: Differences in media appear to remarkably impact osteoblast gene expression and mineralization. These findings may help improve biomaterial evaluation when transitioning from in vitro assessments to in vivo evaluations. Moreover, our results suggest the possibility that gene expression differences observed in MC3T3-E1 cells reflect the diverse bone formation processes in vivo. Focusing on these genes could facilitate the development of screening methods for bone formation, supporting future clinical applications in orthopedics.
Collapse
Affiliation(s)
- Makoto Izumiya
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan; (M.I.); (H.N.); (H.E.); (R.U.); (M.M.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan
| | - Hidehiko Nobuoka
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan; (M.I.); (H.N.); (H.E.); (R.U.); (M.M.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan
| | - Hono Endo
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan; (M.I.); (H.N.); (H.E.); (R.U.); (M.M.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan
| | - Rintaro Ueno
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan; (M.I.); (H.N.); (H.E.); (R.U.); (M.M.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan
| | - Masaki Mimura
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan; (M.I.); (H.N.); (H.E.); (R.U.); (M.M.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan; (M.I.); (H.N.); (H.E.); (R.U.); (M.M.); (N.S.)
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan; (M.I.); (H.N.); (H.E.); (R.U.); (M.M.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan
| |
Collapse
|
3
|
Chouaib B, Desoutter A, Cuisinier F, Collart-Dutilleul PY. Dental Pulp Stem Cell Conditioned Medium Enhance Osteoblastic Differentiation and Bone Regeneration. Stem Cell Rev Rep 2025; 21:477-490. [PMID: 39514179 DOI: 10.1007/s12015-024-10823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Cell-free approaches, utilizing mesenchymal stem cell secretome, have promising prospects in various fields of regenerative medicine. In this study, we examined in vitro and in vivo the potential of dental pulp stem cell-conditioned medium (DPSC-CM) for bone regeneration. METHODS The secretome of undifferentiated stem cells from dental pulp were collected, and the effects of this DPSC-CM were assessed for osteodifferentiation of osteoblast-like cells (MG-63) and osteoblasts deriving from DPSC. Cell proliferation, alkaline phosphatase (ALP) activity, gene expression of Runt-related transcription factor 2 (Runx2), Bone Sialoprotein (BSP), Osteocalcin (OCN), and extracellular matrix mineralization were evaluated. The rat caudal vertebrae critical size defect model was to investigate the effect of DPSC-CM in vivo. RESULTS Results showed that DPSC-CM induced cell growth, and increased ALP activity and the expression of key marker genes at an early stage of osteoblastic differentiation compared to control. A rat bone defect model was used to illustrate the effect of DPSC-CM in vivo. The bone density within the defects were improved using conditioned medium, even though there was no significant difference between the control and DPSC-CM groups. The analysis of DPSC-CM by human growth factor antibody array revealed the presence of several factors involved in osteogenesis. CONCLUSION Taken together, these findings indicate that DPSC-CM is a promising therapeutic candidate for bone regenerative therapy, accelerating the maturation of osteoblastic cells. And even though safety and efficiency of DPSC-CM have to be confirmed in preclinical studies, these results represent a first step toward clinical application.
Collapse
Affiliation(s)
| | | | - Frédéric Cuisinier
- LBN, Univ. Montpellier, Montpellier, France
- Faculty of Dentistry, Univ. Montpellier, Montpellier, France
- Service Odontologie, CHU de Montpellier, Montpellier, France
| | - Pierre-Yves Collart-Dutilleul
- LBN, Univ. Montpellier, Montpellier, France.
- Faculty of Dentistry, Univ. Montpellier, Montpellier, France.
- Service Odontologie, CHU de Montpellier, Montpellier, France.
| |
Collapse
|
4
|
Caronna F, Limem S, Do LDK, Ronan W, Dolan EB. In vitroevaluation of bioabsorbable poly(lactic acid) (PLA) and poly-4-hydroxybutyrate (P4HB) warp-knitted spacer fabric scaffolds for osteogenic differentiation. Biomed Mater 2025; 20:025011. [PMID: 39787697 DOI: 10.1088/1748-605x/ada85d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/09/2025] [Indexed: 01/12/2025]
Abstract
Bioabsorbable textile scaffolds are promising for bone tissue engineering applications. Their tuneable, porous, fibre-based architecture resembles that of native extracellular matrix, and they can sustain tissue growth while being gradually absorbed in the body. In this work, immortalized mouse calvaria preosteoblast MC3T3-E1 cells were culturedin vitroon two warp-knitted bioabsorbable spacer fabric scaffolds made of poly(lactic acid) (PLA) and poly-4-hydroxybutyrate (P4HB), to investigate their osteogenic properties. Scaffold structure and yarn properties were characterized after manufacturing. Cells were seeded on the two scaffolds and treated with osteogenic media for up to 35 days. Both scaffolds supported similar cell growth patterns, featuring a higher cell density on multifilament yarns, which could be beneficial to drive cell proliferation or related phenomena in localized area of the construct. The increase in alkaline phosphatase activity and the calcium deposition observed on some PLA and P4HB scaffolds after 28 and 35 days of culture, confirm their potential to support MC3T3-E1 cells differentiation, however inconsistent mineralization was observed on the scaffolds. Due to their structural and morphological features, ability to support cell attachment and growth, and their limited osteogenic potential, these PLA and P4HB bioabsorbable textile scaffolds are recommended for further investigation for bone tissue engineering applications.
Collapse
Affiliation(s)
- Flavia Caronna
- Biomechanics Research Centre (BMEC), School of Engineering, University of Galway, Galway, Ireland
| | - Skander Limem
- Tepha Medical Inc. (part of Becton & Dickinson), Lexington, MA, United States of America
| | | | - William Ronan
- Biomechanics Research Centre (BMEC), School of Engineering, University of Galway, Galway, Ireland
| | - Eimear B Dolan
- Biomechanics Research Centre (BMEC), School of Engineering, University of Galway, Galway, Ireland
| |
Collapse
|
5
|
Komatsu K, Chao D, Matsuura T, Kido D, Ogawa T. Advancing osseointegration research: A dynamic three-dimensional (3D) in vitro culture model for dental implants. J Dent Sci 2025; 20:350-360. [PMID: 39873044 PMCID: PMC11763192 DOI: 10.1016/j.jds.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/22/2024] [Indexed: 01/30/2025] Open
Abstract
Background/purpose In-vitro studies are essential for understanding cellular responses, but traditional culture systems often neglect the three-dimensional (3D) structure of real implants, leading to limitations in cellular recruitment and behavior largely governed by gravity. The objective of this study was to pioneer a novel 3D dynamic osteoblastic culture system for assessing the biological capabilities of dental implants in a more clinically and physiologically relevant manner. Materials and methods Rat bone marrow-derived osteoblasts were cultured in a 24-well dish with a vertically positioned dental implant. Controlled rotation using a 3D rotator with 3° tilts was applied. Cell attachment, proliferation, and differentiation on implant surfaces were evaluated in response to different surface topographies, physicochemical properties, and local environments. Results Among the tested rotational speeds (0, 10, 30, 50 rpm), optimal osteoblast attachment and proliferation were observed at 30 rpm. A linear correlation was found between cell attachment and rotation speed up to 30 rpm, declining at 50 rpm. Alkaline phosphatase (ALP) activity and mineralized matrix formation were elevated on newly acid-etched, hydrophilic surfaces compared to their 4-week-old hydrophobic surfaces. Sandblasted implants showed higher ALP activity and matrix mineralization. Adding N-acetyl cysteine to the culture medium increased ALP activity and mineralization. Conclusion Osteoblasts successfully attached, proliferated, and mineralized on dental implants in vitro under optimized dynamic conditions. This system differentiated the biological capabilities of implants with varying surface topographies, wettability, and biochemically modulated environments. These findings support developing a 3D dynamic dental implant culture model, advancing osseointegration research and innovating dental implant designs.
Collapse
Affiliation(s)
- Keiji Komatsu
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Denny Chao
- Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Takanori Matsuura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Daisuke Kido
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
| | - Takahiro Ogawa
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA, USA
- Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, CA, USA
| |
Collapse
|
6
|
Bourne LE, Jayash SN, Michels LV, Hopkinson M, Guppy FM, Clarkin CE, Gard P, Brissett N, Staines KA. Sexually dimorphic effects of prenatal alcohol exposure on the murine skeleton. Biol Sex Differ 2024; 15:51. [PMID: 38890762 PMCID: PMC11186175 DOI: 10.1186/s13293-024-00626-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) can result in lifelong disabilities known as foetal alcohol spectrum disorder (FASD) and is associated with childhood growth deficiencies and increased bone fracture risk. However, the effects of PAE on the adult skeleton remain unclear and any potential sexual dimorphism is undetermined. Therefore, we utilised a murine model to examine sex differences with PAE on in vitro bone formation, and in the juvenile and adult skeleton. METHODS Pregnant C57BL/6J female mice received 5% ethanol in their drinking water during gestation. Primary calvarial osteoblasts were isolated from neonatal offspring and mineralised bone nodule formation and gene expression assessed. Skeletal phenotyping of 4- and 12-week-old male and female offspring was conducted by micro-computed tomography (µCT), 3-point bending, growth plate analyses, and histology. RESULTS Osteoblasts from male and female PAE mice displayed reduced bone formation, compared to control (≤ 30%). Vegfa, Vegfb, Bmp6, Tgfbr1, Flt1 and Ahsg were downregulated in PAE male osteoblasts only, whilst Ahsg was upregulated in PAE females. In 12-week-old mice, µCT analysis revealed a sex and exposure interaction across several trabecular bone parameters. PAE was detrimental to the trabecular compartment in male mice compared to control, yet PAE females were unaffected. Both male and female mice had significant reductions in cortical parameters with PAE. Whilst male mice were negatively affected along the tibial length, females were only distally affected. Posterior cortical porosity was increased in PAE females only. Mechanical testing revealed PAE males had significantly reduced bone stiffness compared to controls; maximum load and yield were reduced in both sexes. PAE had no effect on total body weight or tibial bone length in either sex. However, total growth plate width in male PAE mice compared to control was reduced, whilst female PAE mice were unaffected. 4-week-old mice did not display the altered skeletal phenotype with PAE observed in 12-week-old animals. CONCLUSIONS Evidence herein suggests, for the first time, that PAE exerts divergent sex effects on the skeleton, possibly influenced by underlying sex-specific transcriptional mechanisms of osteoblasts. Establishing these sex differences will support future policies and clinical management of FASD.
Collapse
Affiliation(s)
- Lucie E Bourne
- School of Applied Sciences, Centre for Lifelong Health, University of Brighton, Lewes Road, Brighton, BN2 4GT, UK
| | - Soher N Jayash
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Lysanne V Michels
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Mark Hopkinson
- Comparative Biomedical Sciences, Royal Veterinary College, London, NW1 0TU, UK
| | - Fergus M Guppy
- Institute of Life and Earth Sciences, School of Energy, Geosciences, Infrastructure and Society, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Claire E Clarkin
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Paul Gard
- School of Applied Sciences, Centre for Lifelong Health, University of Brighton, Lewes Road, Brighton, BN2 4GT, UK
| | - Nigel Brissett
- School of Applied Sciences, Centre for Lifelong Health, University of Brighton, Lewes Road, Brighton, BN2 4GT, UK
| | - Katherine A Staines
- School of Applied Sciences, Centre for Lifelong Health, University of Brighton, Lewes Road, Brighton, BN2 4GT, UK.
| |
Collapse
|
7
|
Feng S, Feng Z, Wei Y, Zheng X, Deng Z, Liao Z, Jin Y, Chen R, Zhao L. EEF1B2 regulates bone marrow-derived mesenchymal stem cells bone-fat balance via Wnt/β-catenin signaling. Cell Mol Life Sci 2024; 81:260. [PMID: 38878096 PMCID: PMC11335296 DOI: 10.1007/s00018-024-05297-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 02/25/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
The pathological advancement of osteoporosis is caused by the uneven development of bone marrow-derived mesenchymal stem cells (BMSCs) in terms of osteogenesis and adipogenesis. While the role of EEF1B2 in intellectual disability and tumorigenesis is well established, its function in the bone-fat switch of BMSCs is still largely unexplored. During the process of osteogenic differentiation, we observed an increase in the expression of EEF1B2, while a decrease in its expression was noted during adipogenesis. Suppression of EEF1B2 hindered the process of osteogenic differentiation and mineralization while promoting adipogenic differentiation. On the contrary, overexpression of EEF1B2 enhanced osteogenesis and strongly inhibited adipogenesis. Furthermore, the excessive expression of EEF1B2 in the tibias has the potential to mitigate bone loss and decrease marrow adiposity in mice with osteoporosis. In terms of mechanism, the suppression of β-catenin activity occurred when EEF1B2 function was suppressed during osteogenesis. Our collective findings indicate that EEF1B2 functions as a regulator, influencing the differentiation of BMSCs and maintaining a balance between bone and fat. Our finding highlights its potential as a therapeutic target for diseases related to bone metabolism.
Collapse
Affiliation(s)
- Shuhao Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Zihang Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Yiran Wei
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Xiaoyong Zheng
- Orthopaedic Department, The 4th medical center of Chinese PLA General Hospital, Beijing, 100089, China
| | - Zhonghao Deng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Zheting Liao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Yangchen Jin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Ruge Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Liang Zhao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
8
|
Frost K, Lewis JW, Jones SW, Edwards JR, Naylor AJ, McGettrick HM. The Species Effect: Differential Sphingosine-1-Phosphate Responses in the Bone in Human Versus Mouse. Int J Mol Sci 2024; 25:5118. [PMID: 38791156 PMCID: PMC11121697 DOI: 10.3390/ijms25105118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
The deterioration of osteoblast-led bone formation and the upregulation of osteoclast-regulated bone resorption are the primary causes of bone diseases, including osteoporosis. Numerous circulating factors play a role in bone homeostasis by regulating osteoblast and osteoclast activity, including the sphingolipid-sphingosine-1-phosphate (S1P). However, to date no comprehensive studies have investigated the impact of S1P activity on human and murine osteoblasts and osteoclasts. We observed species-specific responses to S1P in both osteoblasts and osteoclasts, where S1P stimulated human osteoblast mineralisation and reduced human pre-osteoclast differentiation and mineral resorption, thereby favouring bone formation. The opposite was true for murine osteoblasts and osteoclasts, resulting in more mineral resorption and less mineral deposition. Species-specific differences in osteoblast responses to S1P were potentially explained by differential expression of S1P receptor 1. By contrast, human and murine osteoclasts expressed comparable levels of S1P receptors but showed differential expression patterns of the two sphingosine kinase enzymes responsible for S1P production. Ultimately, we reveal that murine models may not accurately represent how human bone cells will respond to S1P, and thus are not a suitable model for exploring S1P physiology or potential therapeutic agents.
Collapse
Affiliation(s)
- Kathryn Frost
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - Jonathan W. Lewis
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - Simon W. Jones
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - James R. Edwards
- Botnar Research Centre, University of Oxford, Oxford OX3 7LD, UK;
| | - Amy J. Naylor
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| | - Helen M. McGettrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (K.F.); (J.W.L.); (S.W.J.); (A.J.N.)
| |
Collapse
|
9
|
Semicheva A, Ersoy U, Vasilaki A, Myrtziou I, Kanakis I. Defining the Most Potent Osteoinductive Culture Conditions for MC3T3-E1 Cells Reveals No Implication of Oxidative Stress or Energy Metabolism. Int J Mol Sci 2024; 25:4180. [PMID: 38673767 PMCID: PMC11050066 DOI: 10.3390/ijms25084180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/26/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The MC3T3-E1 preosteoblastic cell line is widely utilised as a reliable in vitro system to assess bone formation. However, the experimental growth conditions for these cells hugely diverge, and, particularly, the osteogenic medium (OSM)'s composition varies in research studies. Therefore, we aimed to define the ideal culture conditions for MC3T3-E1 subclone 4 cells with regard to their mineralization capacity and explore if oxidative stress or the cellular metabolism processes are implicated. Cells were treated with nine different combinations of long-lasting ascorbate (Asc) and β-glycerophosphate (βGP), and osteogenesis/calcification was evaluated at three different time-points by qPCR, Western blotting, and bone nodule staining. Key molecules of the oxidative and metabolic pathways were also assessed. It was found that sufficient mineral deposition was achieved only in the 150 μg.mL-1/2 mM Asc/βGP combination on day 21 in OSM, and this was supported by Runx2, Alpl, Bglap, and Col1a1 expression level increases. NOX2 and SOD2 as well as PGC1α and Tfam were also monitored as indicators of redox and metabolic processes, respectively, where no differences were observed. Elevation in OCN protein levels and ALP activity showed that mineralisation comes as a result of these differences. This work defines the most appropriate culture conditions for MC3T3-E1 cells and could be used by other research laboratories in this field.
Collapse
Affiliation(s)
- Alexandra Semicheva
- Chester Medical School, Faculty of Health, Medicine and Society, University of Chester, Chester CH1 4BJ, UK; (A.S.); (I.M.)
| | - Ufuk Ersoy
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK; (U.E.); (A.V.)
| | - Aphrodite Vasilaki
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK; (U.E.); (A.V.)
| | - Ioanna Myrtziou
- Chester Medical School, Faculty of Health, Medicine and Society, University of Chester, Chester CH1 4BJ, UK; (A.S.); (I.M.)
| | - Ioannis Kanakis
- Chester Medical School, Faculty of Health, Medicine and Society, University of Chester, Chester CH1 4BJ, UK; (A.S.); (I.M.)
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences (ILCaMS), University of Liverpool, Liverpool L7 8TX, UK; (U.E.); (A.V.)
| |
Collapse
|
10
|
Shaikh S, Baniasadi H, Mehrotra S, Ghosh R, Singh P, Seppälä JV, Kumar A. Strontium-Substituted Nanohydroxyapatite-Incorporated Poly(lactic acid) Composites for Orthopedic Applications: Bioactive, Machinable, and High-Strength Properties. Biomacromolecules 2023; 24:4901-4914. [PMID: 37874127 DOI: 10.1021/acs.biomac.3c00610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Traditional metal-alloy bone fixation devices provide structural support for bone repair but have limitations in actively promoting bone healing and often require additional surgeries for implant removal. In this study, we focused on addressing these challenges by fabricating biodegradable composites using poly(lactic acid) (PLA) and strontium-substituted nanohydroxyapatite (SrHAP) via melt compounding and injection molding. Various percentages of SrHAP (5, 10, 20, and 30% w/w) were incorporated into the PLA matrix. We systematically investigated the structural, morphological, thermal, mechanical, rheological, and dynamic mechanical properties of the prepared composites. Notably, the tensile modulus, a critical parameter for orthopedic implants, significantly improved from 2.77 GPa in pristine PLA to 3.73 GPa in the composite containing 10% w/w SrHAP. The incorporation of SrHAP (10% w/w) into the PLA matrix led to an increased storage modulus, indicating a uniform dispersion of SrHAP within the PLA and good compatibility between the polymer and nanoparticles. Moreover, we successfully fabricated screws using PLA composites with 10% (w/w) SrHAP, demonstrating their formability at room temperature and radiopacity when observed under X-ray microtomography (micro-CT). Furthermore, the water contact angle decreased from 93 ± 2° for pristine PLA to 75 ± 3° for the composite containing SrHAP, indicating better surface wettability. To assess the biological behavior of the composites, we conducted in vitro cell-material tests, which confirmed their osteoconductive and osteoinductive properties. These findings highlight the potential of our developed PLA/SrHAP10 (10% w/w) composites as machinable implant materials for orthopedic applications. In conclusion, our study presents the fabrication and comprehensive characterization of biodegradable composites comprising PLA and strontium-substituted nanohydroxyapatite (SrHAP). These composites exhibit improved mechanical properties, formability, and radiopacity while also demonstrating desirable biological behavior. Our results suggest that these PLA/SrHAP10 composites hold promise as machinable implant materials for orthopedic applications.
Collapse
Affiliation(s)
- Shazia Shaikh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Hossein Baniasadi
- Polymer Technology, School of Chemical Engineering, Aalto University, P.O. Box 16100, Espoo, FI-00076 Aalto, Finland
| | - Shreya Mehrotra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Rupita Ghosh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Prerna Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Jukka V Seppälä
- Polymer Technology, School of Chemical Engineering, Aalto University, P.O. Box 16100, Espoo, FI-00076 Aalto, Finland
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- The Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
- Center of Excellence for Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| |
Collapse
|
11
|
Bourne LE, Davies BK, Millan JL, Arnett TR, Wheeler-Jones CPD, Keen JAC, Roberts SJ, Orriss IR. Evidence that pyrophosphate acts as an extracellular signalling molecule to exert direct functional effects in primary cultures of osteoblasts and osteoclasts. Bone 2023; 176:116868. [PMID: 37549801 DOI: 10.1016/j.bone.2023.116868] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Extracellular pyrophosphate (PPi) is well known for its fundamental role as a physiochemical mineralisation inhibitor. However, information about its direct actions on bone cells remains limited. This study shows that PPi decreased osteoclast formation and resorptive activity by ≤50 %. These inhibitory actions were associated with reduced expression of genes involved in osteoclastogenesis (Tnfrsf11a, Dcstamp) and bone resorption (Ctsk, Car2, Acp5). In osteoblasts, PPi present for the entire (0-21 days) or latter stages of culture (7-21/14-21 days) decreased bone mineralisation by ≤95 %. However, PPi present for the differentiation phase only (0-7/0-14 days) increased bone formation (≤70 %). Prolonged treatment with PPi resulted in earlier matrix deposition and increased soluble collagen levels (≤2.3-fold). Expression of osteoblast (RUNX2, Bglap) and early osteocyte (E11, Dmp1) genes along with mineralisation inhibitors (Spp1, Mgp) was increased by PPi (≤3-fold). PPi levels are regulated by tissue non-specific alkaline phosphatase (TNAP) and ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1). PPi reduced NPP1 expression in both cell types whereas TNAP expression (≤2.5-fold) and activity (≤35 %) were increased in osteoblasts. Breakdown of extracellular ATP by NPP1 represents a key source of PPi. ATP release from osteoclasts and osteoblasts was decreased ≤60 % by PPi and by a selective TNAP inhibitor (CAS496014-12-2). Pertussis toxin, which prevents Gαi subunit activation, was used to investigate whether G-protein coupled receptor (GPCR) signalling mediates the effects of PPi. The actions of PPi on bone mineralisation, collagen production, ATP release, gene/protein expression and osteoclast formation were abolished or attenuated by pertussis toxin. Together these findings show that PPi, modulates differentiation, function and gene expression in osteoblasts and osteoclasts. The ability of PPi to alter ATP release and NPP1/TNAP expression and activity indicates that cells can detect PPi levels and respond accordingly. Our data also raise the possibility that some actions of PPi on bone cells could be mediated by a Gαi-linked GPCR.
Collapse
Affiliation(s)
- Lucie E Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; School of Applied Sciences, University of Brighton, UK
| | - Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; Clinical and Experimental Endocrinology, KU, Leuven, Belgium
| | - Jose Luis Millan
- Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Timothy R Arnett
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; Department of Cell and Developmental Biology, University College London, London, UK
| | | | - Jacob A C Keen
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Scott J Roberts
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| |
Collapse
|
12
|
Mertz EL, Makareeva E, Mirigian LS, Leikin S. Bone Formation in 2D Culture of Primary Cells. JBMR Plus 2022; 7:e10701. [PMID: 36699640 PMCID: PMC9850442 DOI: 10.1002/jbm4.10701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/15/2022] [Accepted: 10/16/2022] [Indexed: 11/13/2022] Open
Abstract
Relevance of mineralized nodules in two-dimensional (2D) osteoblast/osteocyte cultures to bone biology, pathology, and engineering is a decades old question, but a comprehensive answer appears to be still wanting. Bone-like cells, extracellular matrix (ECM), and mineral were all reported but so were non-bone-like ones. Many studies described seemingly bone-like cell-ECM structures based on similarity to few select bone features in vivo, yet no studies examined multiple bone features simultaneously and none systematically studied all types of structures coexisting in the same culture. Here, we report such comprehensive analysis of 2D cultures based on light and electron microscopies, Raman microspectroscopy, gene expression, and in situ messenger RNA (mRNA) hybridization. We demonstrate that 2D cultures of primary cells from mouse calvaria do form bona fide bone. Cells, ECM, and mineral within it exhibit morphology, structure, ultrastructure, composition, spatial-temporal gene expression pattern, and growth consistent with intramembranous ossification. However, this bone is just one of at least five different types of cell-ECM structures coexisting in the same 2D culture, which vary widely in their resemblance to bone and ability to mineralize. We show that the other two mineralizing structures may represent abnormal (disrupted) bone and cartilage-like structure with chondrocyte-to-osteoblast transdifferentiation. The two nonmineralizing cell-ECM structures may mimic periosteal cambium and pathological, nonmineralizing osteoid. Importantly, the most commonly used culture conditions (10mM β-glycerophosphate) induce artificial mineralization of all cell-ECM structures, which then become barely distinguishable. We therefore discuss conditions and approaches promoting formation of bona fide bone and simple means for distinguishing it from the other cell-ECM structures. Our findings may improve osteoblast differentiation and function analyses based on 2D cultures and extend applications of these cultures to general bone biology and tissue engineering research. Published 2022. This article is a U.S. Government work and is in the public domain in the USA. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Edward L. Mertz
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Elena Makareeva
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Lynn S. Mirigian
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| | - Sergey Leikin
- Eunice Kennedy Shriver National Institute of Health and Human DevelopmentNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
13
|
Hepp N, Frederiksen AL, Duno M, Jørgensen NR, Jensen JEB. Biochemical and clinical manifestations in adults with hypophosphatasia: a national cross-sectional study. Osteoporos Int 2022; 33:2595-2605. [PMID: 35986118 DOI: 10.1007/s00198-022-06536-2] [Citation(s) in RCA: 9] [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] [Received: 01/06/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022]
Abstract
UNLABELLED Hypophosphatasia (HPP) is a rare disease affecting bone mineralization. Adults with HPP have an increased occurrence of low-energy fractures, which cannot be explained by reduced bone mass assessed by dual energy X-ray absorptiometry. The bone phenotype in adults with HPP requires further studies investigating bone strength and bone structural parameters. INTRODUCTION Hypophosphatasia (HPP) is a rare inherited disorder of bone and mineral metabolism, characterized by broad-ranging clinical manifestations and severity. However, studies investigating the clinical spectrum in adults with HPP compared to a control group are scarce. The aim of this study was to evaluate biochemical and clinical characteristics as well as bone health in a Danish cohort of adults with HPP. METHODS We conducted a cross-sectional study assessing biochemical parameters, fracture prevalence, bone mineral density (BMD), bone turnover markers, physical performance and pain characteristics in 40 adults with HPP and 40 sex-, age-, BMI- and menopausal status-matched healthy controls. RESULTS Patients with HPP had a significantly higher prevalence of non-vertebral, low-energy fractures (p = < 0.001). BMD at the lumbar spine, total hip, femoral neck, forearm and whole body did not differ between the groups. Low levels of the bone-specific alkaline phosphatase correlated significantly with higher BMD at the lumbar spine and femoral neck in both groups. The bone formation marker N-terminal propeptide of type 1 procollagen was significantly lower in patients with HPP than healthy controls (p = 0.006). Adults with HPP had significantly reduced walking capability (p = < 0.001) and lower body strength (p = < 0.001). Chronic pain was significantly more prevalent in adults with HPP than the control group (p = 0.029). CONCLUSIONS The increased occurrence of low-energy fractures in adults with HPP is not explained by low BMD. Adults with HPP have reduced physical performance when compared with healthy controls.
Collapse
Affiliation(s)
- Nicola Hepp
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Kettegaard Alle 30, 2650, Hvidovre, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3 B, 2200, Copenhagen, Denmark.
| | - Anja Lisbeth Frederiksen
- Department of Clinical Genetics, Aalborg University Hospital, Ladegaardsgade 5, 9000, Aalborg C, Denmark
- Department of Clinical Research, Aalborg University, Fredrik Bajers Vej 7K, 9220, Aalborg Ø, Denmark
| | - Morten Duno
- Department of Clinical Genetics, University Hospital Copenhagen Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Niklas Rye Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3 B, 2200, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Valdemar Hansens Vej 13, 2600, Glostrup, Denmark
| | - Jens-Erik Beck Jensen
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Kettegaard Alle 30, 2650, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3 B, 2200, Copenhagen, Denmark
| |
Collapse
|
14
|
Tsuji N, Sakamoto T, Hoshi K, Hikita A. Spatiotemporal Analysis of Osteoblast Morphology and Wnt Signal‐Induced Osteoblast Reactivation during Bone Modeling in Vitro. JBMR Plus 2022; 6:e10689. [PMID: 36398107 PMCID: PMC9664540 DOI: 10.1002/jbm4.10689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
Bone nodule formation by differentiating osteoblasts is considered an in vitro model that mimics bone modeling. However, the details of osteoblast behavior and matrix production during bone nodule formation are poorly understood. Here, we present a spatiotemporal analysis system for evaluating osteoblast morphology and matrix production during bone modeling in vitro via two-photon microscopy. Using this system, a change in osteoblast morphology from cuboidal to flat was observed during the formation of mineralized nodules, and this change was quantified. Areas with high bone formation were densely populated with cuboidal osteoblasts, which were characterized by blebs, protruding structures on their cell membranes. Cuboidal osteoblasts with blebs were highly mobile, and osteoblast blebs exhibited a polar distribution. Furthermore, mimicking romosozumab treatment, when differentiated flattened osteoblasts were stimulated with BIO, a GSK3β inhibitor, they were reactivated to acquire a cuboidal morphology with blebs on their membranes and produced more matrix than nonstimulated cells. Our analysis system is a powerful tool for evaluating the cell morphology and function of osteoblasts during bone modeling. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Naoki Tsuji
- Department of Sensory and Motor System Medicine, Graduate School of Medicine The University of Tokyo Tokyo Japan
| | - Tomoaki Sakamoto
- Department of Tissue Engineering The University of Tokyo Hospital Tokyo Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine The University of Tokyo Tokyo Japan
- Department of Tissue Engineering The University of Tokyo Hospital Tokyo Japan
| | - Atsuhiko Hikita
- Department of Tissue Engineering The University of Tokyo Hospital Tokyo Japan
| |
Collapse
|
15
|
Associated changes in stiffness of collagen scaffolds during osteoblast mineralisation and bone formation. BMC Res Notes 2022; 15:310. [PMID: 36153566 PMCID: PMC9509582 DOI: 10.1186/s13104-022-06203-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Engineering bone in 3D is important for both regenerative medicine purposes and for the development of accurate in vitro models of bone tissue. The changing material stiffness of bone tissue had not yet been monitored throughout the process of mineralisation and bone nodule formation by osteoblasts either during in vitro engineering or in development perspective. Results Within this short research note, stiffness changes (Young’s modulus) during in vitro bone formation by primary osteoblasts in dense collagen scaffolds were monitored using atomic force microscopy. Data analysis revealed significant stiffening of 3D bone cultures at day 5 and 8 that was correlated with the onset of mineral deposition (p < 0.00005).
Collapse
|
16
|
Rezaei A, Li Y, Turmaine M, Bertazzo S, Howard CA, Arnett TR, Shakib K, Jell G. Hypoxia mimetics restore bone biomineralisation in hyperglycaemic environments. Sci Rep 2022; 12:13944. [PMID: 35977987 PMCID: PMC9385857 DOI: 10.1038/s41598-022-18067-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/04/2022] [Indexed: 11/08/2022] Open
Abstract
Diabetic patients have an increased risk of fracture and an increased occurrence of impaired fracture healing. Diabetic and hyperglycaemic conditions have been shown to impair the cellular response to hypoxia, via an inhibited hypoxia inducible factor (HIF)-1α pathway. We investigated, using an in vitro hyperglycaemia bone tissue engineering model (and a multidisciplinary bone characterisation approach), the differing effects of glucose levels, hypoxia and chemicals known to stabilise HIF-1α (CoCl2 and DMOG) on bone formation. Hypoxia (1% O2) inhibited bone nodule formation and resulted in discrete biomineralisation as opposed to the mineralised extracellular collagen fibres found in normoxia (20% O2). Unlike hypoxia, the use of hypoxia mimetics did not prevent nodule formation in normal glucose level. Hyperglycaemic conditions (25 mM and 50 mM glucose) inhibited biomineralisation. Interestingly, both hypoxia mimetics (CoCl2 and DMOG) partly restored hyperglycaemia inhibited bone nodule formation. These results highlight the difference in osteoblast responses between hypoxia mimetics and actual hypoxia and suggests a role of HIF-1α stabilisation in bone biomineralisation that extends that of promoting neovascularisation, or other system effects associated with hypoxia and bone regeneration in vivo. This study demonstrates that targeting the HIF pathway may represent a promising strategy for bone regeneration in diabetic patients.
Collapse
Affiliation(s)
- Azadeh Rezaei
- Division of Surgery & Interventional Science, University College London, 9th Floor Royal Free Hospital, London, NW3 2QG, UK
| | - Yutong Li
- Division of Surgery & Interventional Science, University College London, 9th Floor Royal Free Hospital, London, NW3 2QG, UK
| | - Mark Turmaine
- Department of Cell & Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Sergio Bertazzo
- Department of Medical Physics & Biomedical Engineering, University College London, London, WC1E 6BT, UK
| | - Christopher A Howard
- Department of Physics & Astronomy, University College London, London, WC1E 6BT, UK
| | - Timothy R Arnett
- Department of Cell & Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Kaveh Shakib
- Division of Surgery & Interventional Science, University College London, 9th Floor Royal Free Hospital, London, NW3 2QG, UK.
| | - Gavin Jell
- Division of Surgery & Interventional Science, University College London, 9th Floor Royal Free Hospital, London, NW3 2QG, UK.
| |
Collapse
|
17
|
The Effect of Osteoblast Isolation Methods from Adult Rats on Osteoclastogenesis in Co-Cultures. Int J Mol Sci 2022; 23:ijms23147875. [PMID: 35887222 PMCID: PMC9318333 DOI: 10.3390/ijms23147875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/04/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Co-cultures of osteoblasts and osteoclasts are on the rise because they enable a more complex study. Diseases such as osteoporosis are related to a higher age. Thus, cell isolation from adult individuals is necessary. Osteoblasts can be isolated from the rat femur by three methods: explant culture, explant culture with enzymatic pre-treatment, or enzymatic treatment. The isolation methods yield different populations of osteoblasts which, in a co-culture with peripheral blood mononuclear cells, might result in differences in osteoclastogenesis. Therefore, we examined the differences in osteogenic markers, cell proliferation, and the metabolic activity of isolated osteoblast-like cells in a growth and differentiation medium. We then evaluated the effect of the isolated populations of osteoblast-like cells on osteoclastogenesis in a subsequent co-culture by evaluating osteoclast markers, counting formed osteoclast-like cells, and analyzing their area and number of nuclei. Co-cultures were performed in the presence or absence of osteoclastogenic growth factors, M-CSF and RANKL. It was discovered that enzymatic isolation is not feasible in adult rats, but explant culture and explant culture with enzymatic pre-treatment were both successful. Explant culture with enzymatic pre-treatment yielded cells with a higher proliferation than explant culture in a growth medium. The differentiation medium reduced differences in proliferation during the culture. Some differences in metabolic activity and ALP activity were also found between the osteoblast-like cells isolated by explant culture or by explant culture with enzymatic pre-treatment, but only on some days of cultivation. According to microscopy, the presence of exogenous growth factors supporting osteoclastogenesis in co-cultures was necessary for the formation of osteoclast-like cells. In this case, the formation of a higher number of osteoclast-like cells with a larger area was observed in the co-culture with osteoblast-like cells isolated by explant culture compared to the explant culture with enzymatic pre-treatment. Apart from this observation, no differences in osteoclast markers were noted between the co-cultures with osteoblast-like cells isolated by explant culture and the explant culture with enzymatic pre-treatment. The TRAP and CA II activity was higher in the co-cultures with exogenous growth than that in the co-cultures without exogenous growth factors on day 7, but the opposite was true on day 14. To conclude, explant culture and explant culture with enzymatic pre-treatment are both suitable methods to yield osteoblast-like cells from adult rats capable of promoting osteoclastogenesis in a direct co-culture with peripheral blood mononuclear cells. Explant culture with enzymatic pre-treatment yielded cells with a higher proliferation. The explant culture yielded osteoblast-like cells which induced the formation of a higher number of osteoclast-like cells with a larger area compared to the explant culture with enzymatic pre-treatment when cultured with exogenous M-CSF and RANKL.
Collapse
|
18
|
Tanaka M, Izumiya M, Haniu H, Ueda K, Ma C, Ueshiba K, Ideta H, Sobajima A, Uchiyama S, Takahashi J, Saito N. Current Methods in the Study of Nanomaterials for Bone Regeneration. NANOMATERIALS 2022; 12:nano12071195. [PMID: 35407313 PMCID: PMC9000656 DOI: 10.3390/nano12071195] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/18/2022]
Abstract
Nanomaterials show great promise as bone regeneration materials. They can be used as fillers to strengthen bone regeneration scaffolds, or employed in their natural form as carriers for drug delivery systems. A variety of experiments have been conducted to evaluate the osteogenic potential of bone regeneration materials. In vivo, such materials are commonly tested in animal bone defect models to assess their bone regeneration potential. From an ethical standpoint, however, animal experiments should be minimized. A standardized in vitro strategy for this purpose is desirable, but at present, the results of studies conducted under a wide variety of conditions have all been evaluated equally. This review will first briefly introduce several bone regeneration reports on nanomaterials and the nanosize-derived caveats of evaluations in such studies. Then, experimental techniques (in vivo and in vitro), types of cells, culture media, fetal bovine serum, and additives will be described, with specific examples of the risks of various culture conditions leading to erroneous conclusions in biomaterial analysis. We hope that this review will create a better understanding of the evaluation of biomaterials, including nanomaterials for bone regeneration, and lead to the development of versatile assessment methods that can be widely used in biomaterial development.
Collapse
Affiliation(s)
- Manabu Tanaka
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Makoto Izumiya
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Katsuya Ueda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Chuang Ma
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Koki Ueshiba
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Hirokazu Ideta
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Atsushi Sobajima
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
- Department of Orthopedics (Lower Limbs), Social Medical Care Corporation Hosei-kai Marunouchi Hospital, 1-7-45 Nagisa, Matsumoto, Nagano 390-8601, Japan
| | - Shigeharu Uchiyama
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
| | - Jun Takahashi
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
| |
Collapse
|
19
|
Abidin IZZ, Manogaran T, Wahab RMA, Yazid F, Ariffin SHZ. A Comparative Analysis of Ascorbic Acid-induced Cytotoxicity and Differentiation between SHED and DPSC. Curr Stem Cell Res Ther 2022; 17:576-588. [DOI: 10.2174/1574888x17666220124141310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/13/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022]
Abstract
Aim:
The aim of this study was to compare dental pulp tissue in human exfoliated deciduous teeth (SHEDs) and dental pulp stem cells (DPSCs) in response to ascorbic acid as the sole osteoblast inducer.
Background:
A cocktail of ascorbic acid, β-glycerophosphate, and dexamethasone has been widely used to induce osteoblast differentiation. However, under certain conditions, β-glycerophosphate and dexamethasone can cause a decrease in cell viability in stem cells.
Objectives:
This study aims to determine the cytotoxic effect and potential of ascorbic acid as the sole inducer of osteoblast differentiation.
Methods:
Cytotoxicity analyses in the presence of 10-500 µg/mL ascorbic acid were performed in both cell types using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The concentrations below the IC50 (i.e., 10-150 µg/mL) were used to determine osteoblast differentiation potential of ascorbic acid using the alkaline phosphatase (ALP) assay, von Kossa staining, and reverse transcription-polymerase chain reaction.
Results:
SHEDs and DPSCs proliferated for 21 days, expressed a Mesenchymal Stem Cell (MSC) marker (CD73+), and did not express Hematopoietic Stem Cell (HSC) markers (CD34- and SLAMF1-). SHEDs had a higher range of IC50 values (215-240 µg/mL ascorbic acid), while the IC50 values for DPSCs were 177-211 µg/mL after 24-72 hours. SHEDs treated with 10-100 µg/mL ascorbic acid alone exhibited higher ALP-specific activity and a higher percentage of mineralisation than DPSCs. Both cell types expressed osteoblast markers on day 21, i.e., RUNX2+ and BSP+, in the presence of ascorbic acid.
Conclusions:
SHEDs survive at higher concentrations of ascorbic acid as compared to DPSC. The cytotoxic effect was only exhibited at ≥250 µg/mL ascorbic acid. In addition, SHED exhibited better ALP and mineralization activities, but lower osteoblast marker expression than DPSC in response to ascorbic acid as the sole inducer.
Collapse
Affiliation(s)
| | - Thanaletchumi Manogaran
- Department of Biological Science and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia
| | - Rohaya Megat Abdul Wahab
- Centre of Family Dental Health, Faculty of Dentistry, Universiti Kebangsaaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Farinawati Yazid
- Centre of Family Dental Health, Faculty of Dentistry, Universiti Kebangsaaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Shahrul Hisham Zainal Ariffin
- Department of Biological Science and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia
| |
Collapse
|
20
|
Bioengineering the ameloblastoma tumour to study its effect on bone nodule formation. Sci Rep 2021; 11:24088. [PMID: 34916549 PMCID: PMC8677805 DOI: 10.1038/s41598-021-03484-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/03/2021] [Indexed: 01/09/2023] Open
Abstract
Ameloblastoma is a benign, epithelial cancer of the jawbone, which causes bone resorption and disfigurement to patients affected. The interaction of ameloblastoma with its tumour stroma drives invasion and progression. We used stiff collagen matrices to engineer active bone forming stroma, to probe the interaction of ameloblastoma with its native tumour bone microenvironment. This bone-stroma was assessed by nano-CT, transmission electron microscopy (TEM), Raman spectroscopy and gene analysis. Furthermore, we investigated gene correlation between bone forming 3D bone stroma and ameloblastoma introduced 3D bone stroma. Ameloblastoma cells increased expression of MMP-2 and -9 and RANK temporally in 3D compared to 2D. Our 3D biomimetic model formed bone nodules of an average surface area of 0.1 mm2 and average height of 92.37 [Formula: see text] 7.96 μm over 21 days. We demonstrate a woven bone phenotype with distinct mineral and matrix components and increased expression of bone formation genes in our engineered bone. Introducing ameloblastoma to the bone stroma, completely inhibited bone formation, in a spatially specific manner. Multivariate gene analysis showed that ameloblastoma cells downregulate bone formation genes such as RUNX2. Through the development of a comprehensive bone stroma, we show that an ameloblastoma tumour mass prevents osteoblasts from forming new bone nodules and severely restricted the growth of existing bone nodules. We have identified potential pathways for this inhibition. More critically, we present novel findings on the interaction of stromal osteoblasts with ameloblastoma.
Collapse
|
21
|
Misra BB, Jayapalan S, Richards AK, Helderman RCM, Rendina-Ruedy E. Untargeted metabolomics in primary murine bone marrow stromal cells reveals distinct profile throughout osteoblast differentiation. Metabolomics 2021; 17:86. [PMID: 34537901 PMCID: PMC8450216 DOI: 10.1007/s11306-021-01829-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 08/17/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Skeletal homeostasis is an exquisitely regulated process most directly influenced by bone resorbing osteoclasts, bone forming osteoblasts, and the mechano-sensing osteocytes. These cells work together to constantly remodel bone as a mechanism to prevent from skeletal fragility. As such, when an individual experiences a disconnect in these tightly coupled processes, fracture incidence increases, such as during ageing, gonadal hormone deficiency, weightlessness, and diabetes. While therapeutic options have significantly aided in the treatment of low bone mineral density (BMD) or osteoporosis, limited options remain for anabolic or bone forming agents. Therefore, it is of interest to continue to understand how osteoblasts regulate their metabolism to support the energy expensive process of bone formation. OBJECTIVE The current project sought to rigorously characterize the distinct metabolic processes and intracellular metabolite profiles in stromal cells throughout osteoblast differentiation using untargeted metabolomics. METHODS Primary, murine bone marrow stromal cells (BMSCs) were characterized throughout osteoblast differentiation using standard staining protocols, Seahorse XFe metabolic flux analyses, and untargeted metabolomics. RESULTS We demonstrate here that the metabolic footprint of stromal cells undergoing osteoblast differentiation are distinct, and while oxidative phosphorylation drives adenosine triphosphate (ATP) generation early in the differentiation process, mature osteoblasts depend on glycolysis. Importantly, the intracellular metabolite profile supports these findings while also suggesting additional pathways critical for proper osteoblast function. CONCLUSION These data are the first of their kind to characterize these metabolites in conjunction with the bioenergetic profile in primary, murine stromal cells throughout osteoblast differentiation and provide provocative targets for future investigation.
Collapse
Affiliation(s)
- Biswapriya B Misra
- , Pine-211, Raintree Park Dwaraka Krishna, Namburu, 522508, Andhra Pradesh, India
| | - Shobana Jayapalan
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alison K Richards
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ron C M Helderman
- Maine Medical Center Research Institute, Scarborough, ME, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth Rendina-Ruedy
- Maine Medical Center Research Institute, Scarborough, ME, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
22
|
Izumiya M, Haniu M, Ueda K, Ishida H, Ma C, Ideta H, Sobajima A, Ueshiba K, Uemura T, Saito N, Haniu H. Evaluation of MC3T3-E1 Cell Osteogenesis in Different Cell Culture Media. Int J Mol Sci 2021; 22:ijms22147752. [PMID: 34299372 PMCID: PMC8304275 DOI: 10.3390/ijms22147752] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 11/16/2022] Open
Abstract
Many biomaterials have been evaluated using cultured cells. In particular, osteoblast-like cells are often used to evaluate the osteocompatibility, hard-tissue-regeneration, osteoconductive, and osteoinductive characteristics of biomaterials. However, the evaluation of biomaterial osteogenesis-inducing capacity using osteoblast-like cells is not standardized; instead, it is performed under laboratory-specific culture conditions with different culture media. However, the effect of different media conditions on bone formation has not been investigated. Here, we aimed to evaluate the osteogenesis of MC3T3-E1 cells, one of the most commonly used osteoblast-like cell lines for osteogenesis evaluation, and assayed cell proliferation, alkaline phosphatase activity, expression of osteoblast markers, and calcification under varying culture media conditions. Furthermore, the various media conditions were tested in uncoated plates and plates coated with collagen type I and poly-L-lysine, highly biocompatible molecules commonly used as pseudobiomaterials. We found that the type of base medium, the presence or absence of vitamin C, and the freshness of the medium may affect biomaterial regeneration. We posit that an in vitro model that recapitulates in vivo bone formation should be established before evaluating biomaterials.
Collapse
Affiliation(s)
- Makoto Izumiya
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Miyu Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
| | - Katsuya Ueda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Haruka Ishida
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Chuang Ma
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Hirokazu Ideta
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopaedic Surgery, School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Atsushi Sobajima
- Department of Orthopaedic Surgery, School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopedics (Lower Limbs), Social Medical Care Corporation Hosei-kai Marunouchi Hospital, 1-7-45 Nagisa, Matsumoto, Nagano 390-8601, Japan
| | - Koki Ueshiba
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Takeshi Uemura
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (M.H.); (K.U.); (H.I.); (C.M.); (K.U.); (T.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Correspondence: ; Tel.: +81-263-37-3555
| |
Collapse
|
23
|
Duangchan T, Tawonsawatruk T, Angsanuntsukh C, Trachoo O, Hongeng S, Kitiyanant N, Supokawej A. Amelioration of osteogenesis in iPSC-derived mesenchymal stem cells from osteogenesis imperfecta patients by endoplasmic reticulum stress inhibitor. Life Sci 2021; 278:119628. [PMID: 34015290 DOI: 10.1016/j.lfs.2021.119628] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022]
Abstract
AIM Osteogenesis imperfecta (OI) is a hereditary connective tissue disorder primarily caused by mutations in COL1A1 or COL1A2, which encode type I collagen. These mutations affect the quantity and/or quality of collagen composition in bones, leading to bone fragility. Currently, there is still a lack of treatment that addresses disease-causing factors due to an insufficient understanding of the pathological mechanisms involved. MAIN METHODS Induced pluripotent stem cells (iPSCs) were generated from OI patients with glycine substitution mutations in COL1A1 and COL1A2 and developed into mesenchymal stem cells (iPS-MSCs). OI-derived iPS-MSCs underwent in vitro osteogenic induction to study cell growth, osteogenic differentiation capacity, mRNA expression of osteogenic and unfolded protein response (UPR) markers and apoptosis. The effects of 4-phenylbutyric acid (4-PBA) were examined after treatment of OI iPS-MSCs during osteogenesis. KEY FINDINGS OI-derived iPS-MSCs exhibited decreased cell growth and impaired osteogenic differentiation and collagen expression. Expression of UPR genes was increased, which led to an increase in apoptotic cell death. 4-PBA treatment decreased apoptotic cells and reduced expression of UPR genes, including HSPA5, XBP1, ATF4, DDIT3, and ATF6. Osteogenic phenotypes, including RUNX2, SPP1, BGLAP, and IBPS expression, as well as calcium mineralization, were also improved. SIGNIFICANCE MSCs differentiated from disease-specific iPSCs have utility as a disease model for identifying disease-specific treatments. In addition, the ER stress-associated UPR could be a pathogenic mechanism associated with OI. Treatment with 4-PBA alleviated OI pathogenesis by attenuating UPR markers and apoptotic cell death.
Collapse
Affiliation(s)
- Thitinat Duangchan
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand
| | - Tulyapruek Tawonsawatruk
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Chanika Angsanuntsukh
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Objoon Trachoo
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Narisorn Kitiyanant
- Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand.
| |
Collapse
|
24
|
Study on Gelatin/Hydroxyapatite/Chitosan Material Modified with Osteoblast for Bone Bioengineering. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2021. [DOI: 10.1007/s13369-021-05577-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
25
|
Sharma A, Goring A, Johnson PB, Emery RJH, Hesse E, Boyde A, Olsen BR, Pitsillides AA, Oreffo ROC, Mahajan S, Clarkin CE. Multiscale molecular profiling of pathological bone resolves sexually dimorphic control of extracellular matrix composition. Dis Model Mech 2021; 14:dmm048116. [PMID: 33563616 PMCID: PMC7988766 DOI: 10.1242/dmm.048116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/21/2021] [Indexed: 11/28/2022] Open
Abstract
Collagen assembly during development is essential for successful matrix mineralisation, which determines bone quality and mechanocompetence. However, the biochemical and structural perturbations that drive pathological skeletal collagen configuration remain unclear. Deletion of vascular endothelial growth factor (VEGF; also known as VEGFA) in bone-forming osteoblasts (OBs) induces sex-specific alterations in extracellular matrix (ECM) conformation and mineralisation coupled to vascular changes, which are augmented in males. Whether this phenotypic dimorphism arises as a result of the divergent control of ECM composition and its subsequent arrangement is unknown and is the focus of this study. Herein, we used murine osteocalcin-specific Vegf knockout (OcnVEGFKO) and performed ex vivo multiscale analysis at the tibiofibular junction of both sexes. Label-free and non-destructive polarisation-resolved second-harmonic generation (p-SHG) microscopy revealed a reduction in collagen fibre number in males following the loss of VEGF, complemented by observable defects in matrix organisation by backscattered electron scanning electron microscopy. This was accompanied by localised divergence in collagen orientation, determined by p-SHG anisotropy measurements, as a result of OcnVEGFKO. Raman spectroscopy confirmed that the effect on collagen was linked to molecular dimorphic VEGF effects on collagen-specific proline and hydroxyproline, and collagen intra-strand stability, in addition to matrix carbonation and mineralisation. Vegf deletion in male and female murine OB cultures in vitro further highlighted divergence in genes regulating local ECM structure, including Adamts2, Spp1, Mmp9 and Lama1. Our results demonstrate the utility of macromolecular imaging and spectroscopic modalities for the detection of collagen arrangement and ECM composition in pathological bone. Linking the sex-specific genetic regulators to matrix signatures could be important for treatment of dimorphic bone disorders that clinically manifest in pathological nano- and macro-level disorganisation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Aikta Sharma
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Alice Goring
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Peter B. Johnson
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Roger J. H. Emery
- Department of Surgery and Cancer, Faculty of Medicine, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Eric Hesse
- Institute of Molecular Musculoskeletal Research, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Munich 80336, Germany
| | - Alan Boyde
- Dental Physical Sciences, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK
| | - Bjorn R. Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Andrew A. Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Richard O. C. Oreffo
- Centre for Human Development, Stem Cell and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Sumeet Mahajan
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Claire E. Clarkin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
26
|
Mitochondrial dysfunction impairs osteogenesis, increases osteoclast activity, and accelerates age related bone loss. Sci Rep 2020; 10:11643. [PMID: 32669663 PMCID: PMC7363892 DOI: 10.1038/s41598-020-68566-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/24/2020] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of declining bone mineral density, a universal feature of ageing, is not fully understood. Somatic mitochondrial DNA (mtDNA) mutations accumulate with age in human tissues and mounting evidence suggests that they may be integral to the ageing process. To explore the potential effects of mtDNA mutations on bone biology, we compared bone microarchitecture and turnover in an ageing series of wild type mice with that of the PolgAmut/mut mitochondrial DNA ‘mutator’ mouse. In vivo analyses showed an age-related loss of bone in both groups of mice; however, it was significantly accelerated in the PolgAmut/mut mice. This accelerated rate of bone loss is associated with significantly reduced bone formation rate, reduced osteoblast population densities, increased osteoclast population densities, and mitochondrial respiratory chain deficiency in osteoblasts and osteoclasts in PolgAmut/mut mice compared with wild-type mice. In vitro assays demonstrated severely impaired mineralised matrix formation and increased osteoclast resorption by PolgAmut/mut cells. Finally, application of an exercise intervention to a subset of PolgAmut/mut mice showed no effect on bone mass or mineralised matrix formation in vitro. Our data demonstrate that mitochondrial dysfunction, a universal feature of human ageing, impairs osteogenesis and is associated with accelerated bone loss.
Collapse
|
27
|
Ibrahim A, Rodriguez-Florez N, Gardner OFW, Zucchelli E, New SEP, Borghi A, Dunaway D, Bulstrode NW, Ferretti P. Three-dimensional environment and vascularization induce osteogenic maturation of human adipose-derived stem cells comparable to that of bone-derived progenitors. Stem Cells Transl Med 2020; 9:1651-1666. [PMID: 32639692 PMCID: PMC7695642 DOI: 10.1002/sctm.19-0207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
While human adipose‐derived stem cells (hADSCs) are known to possess osteogenic differentiation potential, the bone tissues formed are generally considered rudimentary and immature compared with those made by bone‐derived precursor cells such as human bone marrow‐derived mesenchymal stem cells (hBMSCs) and less commonly studied human calvarium osteoprogenitor cells (hOPs). Traditional differentiation protocols have tended to focus on osteoinduction of hADSCs through the addition of osteogenic differentiation media or use of stimulatory bioactive scaffolds which have not resulted in mature bone formation. Here, we tested the hypothesis that by reproducing the physical as well as biochemical bone microenvironment through the use of three‐dimensional (3D) culture and vascularization we could enhance osteogenic maturation in hADSCs. In addition to biomolecular characterization, we performed structural analysis through extracellular collagen alignment and mineral density in our bone tissue engineered samples to evaluate osteogenic maturation. We further compared bone formed by hADSCs, hBMSCs, and hOPs against mature human pediatric calvarial bone, yet not extensively investigated. Although bone generated by all three cell types was still less mature than native pediatric bone, a fibrin‐based 3D microenvironment together with vascularization boosted osteogenic maturation of hADSC making it similar to that of bone‐derived osteoprogenitors. This demonstrates the important role of vascularization and 3D culture in driving osteogenic maturation of cells easily available but constitutively less committed to this lineage and suggests a crucial avenue for recreating the bone microenvironment for tissue engineering of mature craniofacial bone tissues from pediatric hADSCs, as well as hBMSCs and hOPs.
Collapse
Affiliation(s)
- Amel Ibrahim
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Naiara Rodriguez-Florez
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.,TECNUN Escuela de Ingenieros, Universidad de Navarra, San Sebastian, Spain
| | - Oliver F W Gardner
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Eleonora Zucchelli
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sophie E P New
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alessandro Borghi
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - David Dunaway
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Neil W Bulstrode
- Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Patrizia Ferretti
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
28
|
McGregor NE, Poulton IJ, Walker EC, Sims NA. Testing Bone Formation Induction by Calvarial Injection Assay in vivo. Bio Protoc 2020; 10:e3560. [PMID: 33659531 PMCID: PMC7842514 DOI: 10.21769/bioprotoc.3560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/18/2020] [Accepted: 01/19/2020] [Indexed: 11/02/2022] Open
Abstract
Bone formation occurs during embryogenesis, skeletal growth and during the process of skeletal renewal throughout life. In the process of bone formation, osteoblasts lay down a collagen-containing matrix, termed osteoid, which is gradually hardened by incorporation of mineral crystals. Although osteoblasts can be induced to differentiate and to deposit mineral in culture, this system does not always provide results that reflect the ability of agents to stimulate bone formation in vivo. This protocol describes a rapid and reliable method for testing local administration of agents on bone formation in vivo. In this method, mice are injected with the agent of question for 5 successive days. Fluorochrome labels are injected prior to, and after agents used for testing, and samples are collected and analysed by undecalcified bone histology and histomorphometry. This provides a robust method for assessing the ability of agents to stimulate bone formation, and if a short-term modification is used, can also be used for testing gene responses in bone to the same stimuli.
Collapse
Affiliation(s)
- Narelle E McGregor
- Bone Cell Biology and Disease Unit, St. Vincent’s Institute of Medical Research, Melbourne, Australia
| | - Ingrid J Poulton
- Bone Cell Biology and Disease Unit, St. Vincent’s Institute of Medical Research, Melbourne, Australia
| | - Emma C Walker
- Bone Cell Biology and Disease Unit, St. Vincent’s Institute of Medical Research, Melbourne, Australia
| | - Natalie A Sims
- Bone Cell Biology and Disease Unit, St. Vincent’s Institute of Medical Research, Melbourne, Australia
- Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
29
|
Sharma A, Goring A, Staines KA, Emery RJ, Pitsillides AA, Oreffo RO, Mahajan S, Clarkin CE. Raman spectroscopy links differentiating osteoblast matrix signatures to pro-angiogenic potential. Matrix Biol Plus 2020; 5:100018. [PMID: 33543015 PMCID: PMC7852201 DOI: 10.1016/j.mbplus.2019.100018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/21/2019] [Accepted: 10/09/2019] [Indexed: 01/25/2023] Open
Abstract
Mineralization of bone is achieved by the sequential maturation of the immature amorphous calcium phase to mature hydroxyapatite (HA) and is central in the process of bone development and repair. To study normal and dysregulated mineralization in vitro, substrates are often coated with poly-l-lysine (PLL) which facilitates cell attachment. This study has used Raman spectroscopy to investigate the effect of PLL coating on osteoblast (OB) matrix composition during differentiation, with a focus on collagen specific proline and hydroxyproline and precursors of HA. Deconvolution analysis of murine derived long bone OB Raman spectra revealed collagen species were 4.01-fold higher in OBs grown on PLL. Further, an increase of 1.91-fold in immature mineral species (amorphous calcium phosphate) was coupled with a 9.32-fold reduction in mature mineral species (carbonated apatite) on PLL versus controls. These unique low mineral signatures identified in OBs were linked with reduced alkaline phosphatase enzymatic activity, reduced Alizarin Red staining and altered osteogenic gene expression. The promotion of immature mineral species and restriction of mature mineral species of OB grown on PLL were linked to increased cell viability and pro-angiogenic vascular endothelial growth factor (VEGF) production. These results demonstrate the utility of Raman spectroscopy to link distinct matrix signatures with OB maturation and VEGF release. Importantly, Raman spectroscopy could provide a label-free approach to clinically assess the angiogenic potential of bone during fracture repair or degenerative bone loss.
Collapse
Key Words
- ACP, amorphous calcium phosphate
- ALP, tissue non-specific alkaline phosphatase
- CAP, carbonated apatite
- CCEC, collagenase-collagenase-EDTA-collagenase
- ECM, extracellular matrix
- HA, hydroxyapatite
- HBSS, Hank's balanced salt solution
- MV, matrix vesicles
- OB, osteoblast
- OCP, octacalcium phosphate
- Osteoblast mineralization
- PCA, principle component analysis
- PLL, poly-l-lysine
- Poly-l-lysine
- RT-qPCR, reverse transcription-quantiative PCR
- Raman spectroscopy
- VEGF
- VEGF, vascular endothelial growth factor
Collapse
Affiliation(s)
- Aikta Sharma
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| | - Alice Goring
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| | - Katherine A. Staines
- School of Applied Sciences, Sighthill Campus, Edinburgh Napier University, Edinburgh, EH11 4BN, United Kingdom of Great Britain and Northern Ireland
| | - Roger J.H. Emery
- Department of Surgery and Cancer, Faculty of Medicine, St Mary's Campus, Imperial College London, London, W2 1PG, United Kingdom of Great Britain and Northern Ireland
| | - Andrew A. Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW1 0TU, United Kingdom of Great Britain and Northern Ireland
| | - Richard O.C. Oreffo
- Centre for Human Development, Stem Cell and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom of Great Britain and Northern Ireland
| | - Sumeet Mahajan
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| | - Claire E. Clarkin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
30
|
Marino S, Bishop RT, de Ridder D, Delgado-Calle J, Reagan MR. 2D and 3D In Vitro Co-Culture for Cancer and Bone Cell Interaction Studies. Methods Mol Biol 2019; 1914:71-98. [PMID: 30729461 DOI: 10.1007/978-1-4939-8997-3_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Co-culture assays are used to study the mutual interaction between cells in vitro. This chapter describes 2D and 3D co-culture systems used to study cell-cell signaling crosstalk between cancer cells and bone marrow adipocytes, osteoblasts, osteoclasts, and osteocytes. The chapter provides a step-by-step guide to the most commonly used cell culture techniques, functional assays, and gene expression.
Collapse
Affiliation(s)
- Silvia Marino
- Division Hematology Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Ryan T Bishop
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Daniëlle de Ridder
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Jesus Delgado-Calle
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michaela R Reagan
- Center for Molecular Medicine, Maine Medical Centre Research Institute, Scarborough, ME, USA
| |
Collapse
|
31
|
Variable osteogenic performance of MC3T3-E1 subclones impacts their utility as models of osteoblast biology. Sci Rep 2019; 9:8299. [PMID: 31165768 PMCID: PMC6549152 DOI: 10.1038/s41598-019-44575-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
The spontaneously immortalized murine calvarial cell line MC3T3-E1 and its derivative subclones are widely used models of osteoblast biology. Many investigators have reported conflicting data under seemingly similar experimental conditions, though the specific subclone studied is often not specified. The purpose of this study was to directly compare the commercially available MC3T3-E1 subclones 4, 14, and 24 in terms of responsiveness to osteogenic induction media and/or stimulation with rhPTH[1–34]. We assayed osteogenic gene expression, capacity to deposit and mineralize a collagenous matrix, and the expression and signaling function of PTH1R. Our data demonstrate that each subclone bears little functional resemblance to the others, or to primary calvarial osteoblasts. Specifically, whereas subclone 4 is responsive to PTH stimulation and capable of matrix mineralization, subclones 14 and 24 do not faithfully replicate these key aspects of osteoblast biology. Furthermore, little overlap was observed between the gene expression profile of subclone 4 and primary calvarial osteoblasts. Our experience working with these cell lines demonstrates that the MC3T3-E1 derived cell lines are imperfect models of osteoblast biology, and reinforce the importance of clearly articulating selection and reporting of research materials.
Collapse
|
32
|
Onat B, Tunçer S, Ulusan S, Banerjee S, Erel-Göktepe I. Biodegradable polymer promotes osteogenic differentiation in immortalized and primary osteoblast-like cells. ACTA ACUST UNITED AC 2019; 14:045003. [PMID: 30856612 DOI: 10.1088/1748-605x/ab0e92] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Biodegradable polymers have been broadly used as agents that can complex with and deliver osteoinductive agents, but osteoinductivity of the polymers themselves has been rarely studied. Here we report the osteoinductivity of poly(4-hydroxy-L-proline ester) (PHPE), a biodegradable cationic polymer with cell penetrating properties. Under physiological conditions, PHPE degrades into trans-4-hydroxy-L-proline (trans-Hyp), a non-coded amino acid with essential functions in collagen fibril formation and fibril stability. Treatment of SaOS-2 osteoblast-like cells and hFOB 1.19 primary osteoblast cells with PHPE promoted earlier collagen nodule formation and mineralization of the extracellular matrix compared to untreated cells, even when mineralization activators were absent in the growth medium. Our results indicate that PHPE is a potential osteoinductive agent in vitro that can favor bone regeneration. Moreover, this osteoinductive property could be partly attributed to the degradation product trans-Hyp, which could recapitulate some, but not all of the osteogenic activity. The primary findings of this study can be summarized as follows: treatment of cells with PHPE led to (1) the induction of COL1A1 expression, collagen synthesis and secretion in osteoblast-like cells, (2) mineralization of the ECM in both SaOS-2 and hFOB 1.19 primary osteoblasts, and (3) induction of BMP2 gene and protein expression in osteoblast-like cells, which can promote mineralization of the ECM and regeneration of the bone tissue. Our results suggest that PHPE is a non-cytotoxic polymer and can be potentially used to overcome collagenopathies such as osteogenesis imperfecta.
Collapse
Affiliation(s)
- Bora Onat
- Department of Biotechnology, Middle East Technical University, 06800, Cankaya, Ankara, Turkey
| | | | | | | | | |
Collapse
|
33
|
Patel JJ, Bourne LE, Davies BK, Arnett TR, MacRae VE, Wheeler-Jones CP, Orriss IR. Differing calcification processes in cultured vascular smooth muscle cells and osteoblasts. Exp Cell Res 2019; 380:100-113. [PMID: 31004580 PMCID: PMC6520648 DOI: 10.1016/j.yexcr.2019.04.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 11/15/2022]
Abstract
Arterial medial calcification (AMC) is the deposition of calcium phosphate mineral, often as hydroxyapatite, in the medial layer of the arteries. AMC shares some similarities to skeletal mineralisation and has been associated with the transdifferentiation of vascular smooth muscle cells (VSMCs) towards an osteoblast-like phenotype. This study used primary mouse VSMCs and calvarial osteoblasts to directly compare the established and widely used in vitro models of AMC and bone formation. Significant differences were identified between osteoblasts and calcifying VSMCs. First, osteoblasts formed large mineralised bone nodules that were associated with widespread deposition of an extracellular collagenous matrix. In contrast, VSMCs formed small discrete regions of calcification that were not associated with collagen deposition and did not resemble bone. Second, calcifying VSMCs displayed a progressive reduction in cell viability over time (≤7-fold), with a 50% increase in apoptosis, whereas osteoblast and control VSMCs viability remained unchanged. Third, osteoblasts expressed high levels of alkaline phosphatase (TNAP) activity and TNAP inhibition reduced bone formation by to 90%. TNAP activity in calcifying VSMCs was ∼100-fold lower than that of bone-forming osteoblasts and cultures treated with β-glycerophosphate, a TNAP substrate, did not calcify. Furthermore, TNAP inhibition had no effect on VSMC calcification. Although, VSMC calcification was associated with increased mRNA expression of osteoblast-related genes (e.g. Runx2, osterix, osteocalcin, osteopontin), the relative expression of these genes was up to 40-fold lower in calcifying VSMCs versus bone-forming osteoblasts. In summary, calcifying VSMCs in vitro display some limited osteoblast-like characteristics but also differ in several key respects: 1) their inability to form collagen-containing bone; 2) their lack of reliance on TNAP to promote mineral deposition; and, 3) the deleterious effect of calcification on their viability.
Collapse
Affiliation(s)
- Jessal J Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; School of Life & Medical Sciences, University of Hertfordshire, Hatfield, UK
| | - Lucie E Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| |
Collapse
|
34
|
Baumann L, Kauschke V, Vikman A, Dürselen L, Krasteva-Christ G, Kampschulte M, Heiss C, Yee KT, Vetter DE, Lips KS. Deletion of nicotinic acetylcholine receptor alpha9 in mice resulted in altered bone structure. Bone 2019; 120:285-296. [PMID: 30414510 PMCID: PMC6492625 DOI: 10.1016/j.bone.2018.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/17/2018] [Accepted: 11/05/2018] [Indexed: 12/22/2022]
Abstract
Alterations in bone strength and structure were found in knockout (KO) mouse strains with deletion of several acetylcholine receptors. Interestingly, the expression of the nicotinic acetylcholine receptors (nAChR) subunit α10 was down-regulated in osteogenic differentiated mesenchymal stem cells of patients with osteoporosis whereas the expression of subunit α9 was not altered. Since nAChR subunits α9 and α10 are often combined in a functional receptor, we analyzed here the bone of adult female KO mice with single deletion of either nAChR alpha9 (α9KO) or alpha10 (α10KO). Biomechanical testing showed a significant decrease of bending stiffness and maximal breaking force in α9KO compared to their corresponding wild type mice. Furthermore, an increase in trabecular pattern factor (Tb.Pf) and structure model index (SMI) was detected by μCT in α9KO indicating reduced bone mass. On the mRNA level a decrease of Collagen 1α1 and Connexin-43 was measured by real-time RT-PCR in α9KO while no alteration of osteoclast markers was detected in either mouse strain. Using electron microcopy we observed an increase in the number of osteocytes that showed signs of degeneration and cell death in the α9KO compared to their wild type mice, while α10KO showed no differences. In conclusion, we demonstrate alterations in bone strength, structure and bio-marker expression in α9KO mice which imply the induction of osteocyte degeneration. Thus, our data suggest that nAChR containing the α9 subunit might be involved in the homeostasis of osteocytes and therefore in bone mass regulation.
Collapse
Affiliation(s)
- Lisa Baumann
- Experimental Trauma Surgery, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany.
| | - Vivien Kauschke
- Experimental Trauma Surgery, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany.
| | - Anna Vikman
- Institute of Orthopaedic Research and Biomechanics, Center for Trauma Research, Ulm University Medical Centre, Helmholtzstrasse 14, 89081 Ulm, Germany
| | - Lutz Dürselen
- Institute of Orthopaedic Research and Biomechanics, Center for Trauma Research, Ulm University Medical Centre, Helmholtzstrasse 14, 89081 Ulm, Germany.
| | - Gabriela Krasteva-Christ
- Institute of Anatomy and Cell Biology, Saarland University, Kirrberger Straße, 66421 Homburg, Germany.
| | - Marian Kampschulte
- Laboratory of Experimental Radiology, Justus-Liebig-University Giessen, Schubertstrasse 81, 35392 Giessen, Germany.
| | - Christian Heiss
- Department of Trauma, Hand, and Reconstructive Surgery Giessen, University Hospital of Giessen-Marburg, Rudolf-Buchheim-Str. 7, 35392 Giessen, Germany.
| | - Kathleen T Yee
- Department of Neurobiology and Anatomical Sciences, University Mississippi Medical Center, Jackson, MS, USA.
| | - Douglas E Vetter
- Department of Neurobiology and Anatomical Sciences, University Mississippi Medical Center, Jackson, MS, USA.
| | - Katrin Susanne Lips
- Experimental Trauma Surgery, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany.
| |
Collapse
|
35
|
Abstract
This chapter describes the isolation, culture, and staining of osteoblasts. The key advantages of this assay are that it allows direct measurement of bone matrix deposition and mineralization, as well as yielding good quantities of osteoblasts at defined stages of differentiation for molecular and histological analysis. An additional focus of this chapter will be the culture of osteoblasts from less conventional animal species.
Collapse
Affiliation(s)
- Inês P Perpétuo
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Lucie E Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| |
Collapse
|
36
|
Szewczyk PK, Metwally S, Karbowniczek JE, Marzec MM, Stodolak-Zych E, Gruszczyński A, Bernasik A, Stachewicz U. Surface-Potential-Controlled Cell Proliferation and Collagen Mineralization on Electrospun Polyvinylidene Fluoride (PVDF) Fiber Scaffolds for Bone Regeneration. ACS Biomater Sci Eng 2018; 5:582-593. [DOI: 10.1021/acsbiomaterials.8b01108] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
37
|
Onat B, Ozcubukcu S, Banerjee S, Erel-Goktepe I. Osteoconductive layer-by-layer films of Poly(4-hydroxy-L-proline ester) (PHPE) and Tannic acid. Eur Polym J 2018. [DOI: 10.1016/j.eurpolymj.2018.03.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
38
|
Patel JJ, Zhu D, Opdebeeck B, D’Haese P, Millán JL, Bourne LE, Wheeler-Jones CPD, Arnett TR, MacRae VE, Orriss IR. Inhibition of arterial medial calcification and bone mineralization by extracellular nucleotides: The same functional effect mediated by different cellular mechanisms. J Cell Physiol 2018; 233:3230-3243. [PMID: 28976001 PMCID: PMC5792173 DOI: 10.1002/jcp.26166] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/22/2017] [Indexed: 12/30/2022]
Abstract
Arterial medial calcification (AMC) is thought to share some outward similarities to skeletal mineralization and has been associated with the transdifferentiation of vascular smooth muscle cells (VSMCs) to an osteoblast-like phenotype. ATP and UTP have previously been shown to inhibit bone mineralization. This investigation compared the effects of extracellular nucleotides on calcification in VSMCs with those seen in osteoblasts. ATP, UTP and the ubiquitous mineralization inhibitor, pyrophosphate (PPi ), dose dependently inhibited VSMC calcification by ≤85%. Culture of VSMCs in calcifying conditions was associated with an increase in apoptosis; treatment with ATP, UTP, and PPi reduced apoptosis to levels seen in non-calcifying cells. Extracellular nucleotides had no effect on osteoblast viability. Basal alkaline phosphatase (TNAP) activity was over 100-fold higher in osteoblasts than VSMCs. ATP and UTP reduced osteoblast TNAP activity (≤50%) but stimulated VSMC TNAP activity (≤88%). The effects of extracellular nucleotides on VSMC calcification, cell viability and TNAP activity were unchanged by deletion or inhibition of the P2Y2 receptor. Conversely, the actions of ATP/UTP on bone mineralization and TNAP activity were attenuated in osteoblasts lacking the P2Y2 receptor. Ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1) hydrolyses ATP and UTP to produce PPi . In both VSMCs and osteoblasts, deletion of NPP1 blunted the inhibitory effects of extracellular nucleotides suggesting involvement of P2 receptor independent pathways. Our results show that although the overall functional effect of extracellular nucleotides on AMC and bone mineralization is similar there are clear differences in the cellular mechanisms mediating these actions.
Collapse
Affiliation(s)
- JJ Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - D Zhu
- Guangzhou Institute of Cardiovascular Disease, School of Basic Medical Sciences, Guangzhou Medical University, China
| | - B Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - P D’Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - JL Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - LE Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - CPD Wheeler-Jones
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - TR Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - VE MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - IR Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
39
|
Ikpegbu E, Basta L, Clements DN, Fleming R, Vincent TL, Buttle DJ, Pitsillides AA, Staines KA, Farquharson C. FGF-2 promotes osteocyte differentiation through increased E11/podoplanin expression. J Cell Physiol 2018; 233:5334-5347. [PMID: 29215722 PMCID: PMC5900964 DOI: 10.1002/jcp.26345] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/29/2017] [Indexed: 01/15/2023]
Abstract
E11/podoplanin is critical in the early stages of osteoblast‐to‐osteocyte transitions (osteocytogenesis), however, the upstream events which regulate E11 expression are unknown. The aim of this study was to examine the effects of FGF‐2 on E11‐mediated osteocytogenesis and to reveal the nature of the underlying signaling pathways regulating this process. Exposure of MC3T3 osteoblast‐like cells and murine primary osteoblasts to FGF‐2 (10 ng/ml) increased E11 mRNA and protein expression (p < 0.05) after 4, 6, and 24 hr. FGF‐2 induced changes in E11 expression were also accompanied by significant (p < 0.01) increases in Phex and Dmp1 (osteocyte markers) expression and decreases in Col1a1, Postn, Bglap, and Alpl (osteoblast markers) expression. Immunofluorescent microscopy revealed that FGF‐2 stimulated E11 expression, facilitated the translocation of E11 toward the cell membrane, and subsequently promoted the formation of osteocyte‐like dendrites in MC3T3 and primary osteoblasts. siRNA knock down of E11 expression achieved >70% reduction of basal E11 mRNA expression (p < 0.05) and effectively abrogated FGF‐2‐related changes in E11 expression and dendrite formation. FGF‐2 strongly activated the ERK signaling pathway in osteoblast‐like cells but inhibition of this pathway did not block the ability of FGF‐2 to enhance E11 expression or to promote acquisition of the osteocyte phenotype. The results of this study highlight a novel mechanism by which FGF‐2 can regulate osteoblast differentiation and osteocyte formation. Specifically, the data suggests that FGF‐2 promotes osteocytogenesis through increased E11 expression and further studies will identify if this regulatory pathway is essential for bone development and maintenance in health and disease.
Collapse
Affiliation(s)
- Ekele Ikpegbu
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK.,Michael Okpara University of Agriculture, Abia, Nigeria
| | - Lena Basta
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Dylan N Clements
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Robert Fleming
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Tonia L Vincent
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - David J Buttle
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield Medical School, Sheffield, UK
| | | | | | - Colin Farquharson
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
40
|
Calcitonin gene‑related peptide reduces Porphyromonas gingivalis LPS‑induced TNF‑α release and apoptosis in osteoblasts. Mol Med Rep 2017; 17:3246-3254. [PMID: 29257246 DOI: 10.3892/mmr.2017.8205] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 05/23/2017] [Indexed: 11/05/2022] Open
Abstract
Periodontal diseases comprise mixed bacterial infections mainly caused by Gram‑negative anaerobic bacteria. Lipopolysaccharides (LPS) are important virulence factors and periodontal pathogens, which change local cytokine levels and promote osteoblast apoptosis, thereby leading to an imbalance in bone remodeling mechanisms and accelerating bone loss. Calcitonin gene‑related peptide (CGRP) is a vasoactive neuropeptide that is released from sensory nerves and has a positive effect on osteoblast proliferation and differentiation. In addition, this small molecule peptide is an important immune regulator in the inflammatory response. The aim of the present study was to assess the in vitro effects of CGRP on Porphyromonas gingivalis (Pg)LPS‑induced osteoblast apoptosis. Osteoblast cultures were stimulated either with various concentrations of PgLPS (0, 25, 50, 100, 500 and 1,000 ng/ml) for 48 h or with 500 ng/ml PgLPS for various lengths of time (0, 6, 12, 24, 48 and 72 h). The PgLPS‑stimulated cells were pretreated with different concentrations of CGRP (0, 1, 10, 100 and 1,000 nM) and cell viability and apoptotic rates were measured by Cell Counting kit‑8 assays and flow cytometry, respectively. CGRP, cleaved (c)‑Caspase‑8 and c‑Caspase‑3 protein expression levels were analyzed by western blotting. Changes in cytokine expression levels, which included tumor necrosis factor (TNF)‑α, interleukin (IL)‑1β, IL‑6, monocyte chemotactic protein (MCP)‑1 and MCP‑2, were measured by ELISA. PgLPS was demonstrated to inhibit osteoblast viability and promote apoptosis in a time‑ and concentration‑dependent manner. CGRP expression was revealed to reduce PgLPS‑induced cytostatic activity and apoptosis in osteoblasts. CGRP also suppressed the PgLPS‑induced release of TNF‑α and inhibited the activation of c‑Caspase‑3 and c‑Caspase‑8, thus preventing apoptosis in osteoblasts. CGRP may be an important neuropeptide in bone remodeling and may reduce osteoblast apoptosis in inflammatory conditions. These results may provide a solid foundation for CGRP to serve as a new target for the treatment of periodontitis.
Collapse
|
41
|
Smith SJ, Emery R, Pitsillides A, Clarkin CE, Mahajan S. Detection of early osteogenic commitment in primary cells using Raman spectroscopy. Analyst 2017; 142:1962-1973. [DOI: 10.1039/c6an02469f] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Raman spectroscopy as a simple and sensitive method to measure early osteogenic responses in primary cultures of bone cells is presented.
Collapse
Affiliation(s)
| | - Roger Emery
- Division of Surgery
- Reproductive Biology and Anaesthetics
- Imperial College London
- UK
| | | | | | - Sumeet Mahajan
- Department of Chemistry and the Institute for Life Sciences
- University of Southampton
- UK
| |
Collapse
|
42
|
Strain dependent differences in glucocorticoid-induced bone loss between C57BL/6J and CD-1 mice. Sci Rep 2016; 6:36513. [PMID: 27812009 PMCID: PMC5109915 DOI: 10.1038/srep36513] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/30/2016] [Indexed: 01/08/2023] Open
Abstract
We have investigated the effect of long-term glucocorticoid (GC) administration on bone turnover in two frequently used mouse strains; C57BL/6J and CD1, in order to assess the influence of their genetic background on GC-induced osteoporosis (GIO). GIO was induced in 12 weeks old female C57BL/6J and CD1 mice by subcutaneous insertion of long-term release prednisolone or placebo pellets. Biomechanical properties as assessed by three point bent testing revealed that femoral elasticity and strength significantly decreased in CD1 mice receiving GC, whereas C57BL/6J mice showed no differences between placebo and prednisolone treatment. Bone turnover assessed by microcomputer tomography revealed that contrary to C57BL/6J mice, prednisolone treated CD1 mice developed osteoporosis. In vitro experiments have underlined that, at a cellular level, C57BL/6J mice osteoclasts and osteoblasts were less responsive to GC treatment and tolerated higher doses than CD1 cells. Whilst administration of long-term release prednisolone pellets provided a robust GIO animal model in 12 weeks old CD1 mice, age matched C57BL/6J mice were not susceptible to the bone changes associated with GIO. This study indicates that for the induction of experimental GIO, the mouse strain choice together with other factors such as age should be carefully evaluated.
Collapse
|
43
|
Hajjawi MOR, Patel JJ, Corcelli M, Arnett TR, Orriss IR. Lack of effect of adenosine on the function of rodent osteoblasts and osteoclasts in vitro. Purinergic Signal 2016; 12:247-58. [PMID: 26861849 DOI: 10.1007/s11302-016-9499-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/01/2016] [Indexed: 01/06/2023] Open
Abstract
Extracellular ATP, signalling through P2 receptors, exerts well-documented effects on bone cells, inhibiting mineral deposition by osteoblasts and stimulating the formation and resorptive activity of osteoclasts. The aims of this study were to determine the potential osteotropic effects of adenosine, the hydrolysis product of ATP, on primary bone cells in vitro. We determined the effect of exogenous adenosine on (1) the growth, alkaline phosphatase (TNAP) activity and bone-forming ability of osteoblasts derived from the calvariae of neonatal rats and mice and the marrow of juvenile rats and (2) the formation and resorptive activity of osteoclasts from juvenile mouse marrow. Reverse transcription polymerase chain reaction (RT-PCR) analysis showed marked differences in the expression of P1 receptors in osteoblasts from different sources. Whilst mRNA for the A1 and A2B receptors was expressed by all primary osteoblasts, A2A receptor expression was limited to rat bone marrow and mouse calvarial osteoblasts and the A3 receptor to rat bone marrow osteoblasts. We found that adenosine had no detectable effects on cell growth, TNAP activity or bone formation by rodent osteoblasts in vitro. The analogue 2-chloroadenosine, which is hydrolysed more slowly than adenosine, had no effects on rat or mouse calvarial osteoblasts but increased TNAP activity and bone formation by rat bone marrow osteoblasts by 30-50 % at a concentration of 1 μM. Osteoclasts were found to express the A2A, A2B and A3 receptors; however, neither adenosine (≤100 μM) nor 2-chloroadenosine (≤10 μM) had any effect on the formation or resorptive activity of mouse osteoclasts in vitro. These results suggest that adenosine, unlike ATP, is not a major signalling molecule in the bone.
Collapse
Affiliation(s)
- Mark O R Hajjawi
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK
| | - Jessal J Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Michelangelo Corcelli
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK.
| | - Isabel R Orriss
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK.,Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|
44
|
Qiao X, Yong Qiao X, Nie Y, Ma Y, Xian Ma Y, Chen Y, Cheng R, Yin W, Yao Yinrg W, Hu Y, Xu W, Ming Xu W, Xu L, Zhi Xu L. Irisin promotes osteoblast proliferation and differentiation via activating the MAP kinase signaling pathways. Sci Rep 2016; 6:18732. [PMID: 26738434 PMCID: PMC4704023 DOI: 10.1038/srep18732] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/25/2015] [Indexed: 12/25/2022] Open
Abstract
Physical exercise is able to improve skeletal health. However, the mechanisms are poorly known. Irisin, a novel exercise-induced myokine, secreted by skeletal muscle in response to exercise, have been shown to mediate beneficial effects of exercise in many disorders. In the current study, we demonstrated that irisin promotes osteoblast proliferation, and increases the expression of osteoblastic transcription regulators, such as Runt-related transcription factor-2, osterix/sp7; and osteoblast differentiation markers, including alkaline phosphatase, collagen type 1 alpha-1, osteocalcin, and osteopontin in vitro. Irisin also increase ALP activity and calcium deposition in cultured osteoblast. These osteogenic effects were mediated by activating the p38 mitogen-activated protein kinase (p-p38 MAPK) and extracellular signal-regulated kinase (ERK). Inhibition of p38 MAPK by SB023580 or pERK by U0126 abolished the proliferation and up-regulatory effects of irisin on Runx2 expression and ALP activity. Together our observation suggest that irisin directly targets osteoblast, promoting osteoblast proliferation and differentiation via activating P38/ERK MAP kinase signaling cascades in vitro. Whether irisin can be utilized as the therapeutic agents for osteopenia and osteoporosis is worth to be further pursued.
Collapse
Affiliation(s)
| | - Xiao Yong Qiao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | - Ying Nie
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | | | - Ya Xian Ma
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | - Yan Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | - Ran Cheng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | | | - Wei Yao Yinrg
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | - Ying Hu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | | | - Wen Ming Xu
- The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | - Liangzhi Xu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, People's Republic of China
| | | |
Collapse
|
45
|
Staines KA, Prideaux M, Allen S, Buttle DJ, Pitsillides AA, Farquharson C. E11/Podoplanin Protein Stabilization Through Inhibition of the Proteasome Promotes Osteocyte Differentiation in Murine in Vitro Models. J Cell Physiol 2015; 231:1392-404. [PMID: 26639105 PMCID: PMC4832367 DOI: 10.1002/jcp.25282] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/04/2015] [Indexed: 11/08/2022]
Abstract
The transmembrane glycoprotein E11 is considered critical in early osteoblast-osteocyte transitions (osteocytogenesis), however its function and regulatory mechanisms are still unknown. Using the late osteoblast MLO-A5 cell line we reveal increased E11 protein/mRNA expression (P < 0.001) concomitant with extensive osteocyte dendrite formation and matrix mineralization (P < 0.001). Transfection with E11 significantly increased mRNA levels (P < 0.001), but immunoblotting failed to detect any correlative increases in E11 protein levels, suggestive of post-translational degradation. We found that exogenous treatment of MLO-A5 and osteocytic IDG-SW3 cells with 10 μM ALLN (calpain and proteasome inhibitor) stabilized E11 protein levels and induced a profound increase in osteocytic dendrite formation (P < 0.001). Treatment with other calpain inhibitors failed to promote similar osteocytogenic changes, suggesting that these effects of ALLN rely upon its proteasome inhibitor actions. Accordingly we found that proteasome-selective inhibitors (MG132/lactacystin/ Bortezomib/Withaferin-A) produced similar dose-dependent increases in E11 protein levels in MLO-A5 and primary osteoblast cells. This proteasomal targeting was confirmed by immunoprecipitation of ubiquitinylated proteins, which included E11, and by increased levels of ubiquitinylated E11 protein upon addition of the proteasome inhibitors MG132/Bortezomib. Activation of RhoA, the small GTPase, was found to be increased concomitant with the peak in E11 levels and its downstream signaling was also observed to promote MLO-A5 cell dendrite formation. Our data indicate that a mechanism reliant upon blockade of proteasome-mediated E11 destabilization contributes to osteocytogenesis and that this may involve downstream targeting of RhoA. This work adds to our mechanistic understanding of the factors regulating bone homeostasis, which may lead to future therapeutic approaches.
Collapse
Affiliation(s)
- Katherine A Staines
- Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Matt Prideaux
- The University of Adelaide, North Terrace, Adelaide, Australia
| | - Steve Allen
- Royal Veterinary College, Royal College Street, London, United Kingdom
| | - David J Buttle
- Department of Infection and Immunity, The University of Sheffield Medical School, Beech Hill Road, Sheffield, United Kingdom
| | | | - Colin Farquharson
- Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| |
Collapse
|
46
|
Orriss IR, Key ML, Hajjawi MOR, Millán JL, Arnett TR. Acidosis is a key regulator of osteoblast ecto-nucleotidase pyrophosphatase/phosphodiesterase 1 (NPP1) expression and activity. J Cell Physiol 2015; 230:3049-56. [PMID: 26033523 PMCID: PMC4549203 DOI: 10.1002/jcp.25041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/07/2015] [Indexed: 01/19/2023]
Abstract
Previous work has shown that acidosis prevents bone nodule formation by osteoblasts in vitro by inhibiting mineralisation of the collagenous matrix. The ratio of phosphate (Pi) to pyrophosphate (PPi) in the bone microenvironment is a fundamental regulator of bone mineralisation. Both Pi and PPi, a potent inhibitor of mineralisation, are generated from extracellular nucleotides by the actions of ecto‐nucleotidases. This study investigated the expression and activity of ecto‐nucleotidases by osteoblasts under normal and acid conditions. We found that osteoblasts express mRNA for a number of ecto‐nucleotidases including NTPdase 1–6 (ecto‐nucleoside triphosphate diphosphohydrolase) and NPP1‐3 (ecto‐nucleotide pyrophosphatase/phosphodiesterase). The rank order of mRNA expression in differentiating rat osteoblasts (day 7) was Enpp1 > NTPdase 4 > NTPdase 6 > NTPdase 5 > alkaline phosphatase > ecto‐5‐nucleotidase > Enpp3 > NTPdase 1 > NTPdase 3 > Enpp2 > NTPdase 2. Acidosis (pH 6.9) upregulated NPP1 mRNA (2.8‐fold) and protein expression at all stages of osteoblast differentiation compared to physiological pH (pH 7.4); expression of other ecto‐nucleotidases was unaffected. Furthermore, total NPP activity was increased up to 53% in osteoblasts cultured in acid conditions (P < 0.001). Release of ATP, one of the key substrates for NPP1, from osteoblasts, was unaffected by acidosis. Further studies showed that mineralised bone formation by osteoblasts cultured from NPP1 knockout mice was increased compared with wildtypes (2.5‐fold, P < 0.001) and was partially resistant to the inhibitory effect of acidosis. These results indicate that increased NPP1 expression and activity might contribute to the decreased mineralisation observed when osteoblasts are exposed to acid conditions. J. Cell. Physiol. 230: 3049–3056, 2015. © 2015 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.,Department of Cell and Developmental Biology, University College London, London, UK
| | - Michelle L Key
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Mark O R Hajjawi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - José L Millán
- Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
47
|
Hajjawi MOR, MacRae VE, Huesa C, Boyde A, Millán JL, Arnett TR, Orriss IR. Mineralisation of collagen rich soft tissues and osteocyte lacunae in Enpp1(-/-) mice. Bone 2014; 69:139-47. [PMID: 25260930 PMCID: PMC4228085 DOI: 10.1016/j.bone.2014.09.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/26/2014] [Accepted: 09/18/2014] [Indexed: 01/23/2023]
Abstract
Ecto-nucleotide pyrophosphatase/phosphodiesterases (NPPs) hydrolyse nucleotide triphosphates to the corresponding nucleotide monophosphates and the mineralisation inhibitor, pyrophosphate (PPi). This study examined the role of NPP1 in osteocytes, osteoclasts and cortical bone, using a mouse model lacking NPP1 (Enpp1(-/-)). We used microcomputed tomography (μCT) to investigate how NPP1 deletion affects cortical bone structure; excised humerus bones from 8, 15 and 22-week old mice were scanned at 0.9 μm. Although no changes were evident in the cortical bone of 8-week old Enpp1(-/-) mice, significant differences were observed in older animals. Cortical bone volume was decreased 28% in 22-week Enpp1(-/-) mice, whilst cortical porosity was reduced 30% and 60% at 15 and 22-weeks, respectively. This was accompanied by up to a 15% decrease in closed pore diameter and a 55% reduction in the number of pores. Cortical thickness was reduced up to 35% in 15 and 22-week Enpp1(-/-) animals and the endosteal diameter was increased up to 23%. Thus, the cortical bone from Enpp1(-/-) mice was thinner and less porous, with a larger marrow space. Scanning electron microscopy (SEM) revealed a decrease in the size and number of blood vessel channels in the cortical bone as well as a 40% reduction in the mean plan area of osteocyte lacunae. We noted that the number of viable osteocytes isolated from the long bones of Enpp1(-/-) mice was decreased ≤50%. In contrast, osteoclast formation and resorptive activity were unaffected by NPP1 deletion. μCT and histological analysis of Enpp1(-/-) mice also revealed calcification of the joints and vertebrae as well as soft tissues including the whisker follicles, ear pinna and trachea. This calcification worsened as the animals aged. Together, these data highlight the key role of NPP1 in regulating calcification of both soft and skeletal tissues.
Collapse
Affiliation(s)
- Mark O R Hajjawi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Carmen Huesa
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Alan Boyde
- Institute of Dentistry, Bart's and the London School of Medicine and Dentistry, Queen Mary, University of London, UK
| | | | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| |
Collapse
|
48
|
Taylor SEB, Shah M, Orriss IR. Generation of rodent and human osteoblasts. BONEKEY REPORTS 2014; 3:585. [PMID: 25396049 DOI: 10.1038/bonekey.2014.80] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/04/2014] [Indexed: 01/18/2023]
Abstract
This paper describes the isolation, culture and staining of primary osteoblasts from neonatal rodents and human samples. The calvaria and long-bone assays allow direct measurement of bone matrix deposition and mineralisation, as well as producing osteoblasts at defined stages of differentiation for molecular and histological analysis. Culture of human osteoblasts enables cell function to be investigated in targeted patient groups. The described methods will provide a step-by-step guide of what to expect at each stage of the culture and highlight the varied tissue culture conditions required to successfully grow osteoblasts from different sources. A special focus of this paper is the methods used for analysis of bone mineralisation and how to ensure that nonspecific mineral deposition or staining is not quantified.
Collapse
Affiliation(s)
- Sarah E B Taylor
- Department of Orthopaedic Surgery, Stanford University School of Medicine , Stanford, CA, USA
| | - Mittal Shah
- Department of Surgery and Cancer, Imperial College London , London, UK ; Department of Comparative Biomedical Sciences, Royal Veterinary College , London, UK
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College , London, UK
| |
Collapse
|