201
|
The Role of Exosomes in Bone Remodeling: Implications for Bone Physiology and Disease. DISEASE MARKERS 2019; 2019:9417914. [PMID: 31485281 PMCID: PMC6710799 DOI: 10.1155/2019/9417914] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022]
Abstract
Bone remodeling represents a physiological phenomenon of continuous bone tissue renewal that requires fine orchestration of multiple cell types, which is critical for the understanding of bone disease but not yet clarified in precise detail. Exosomes, which are cell-secreted nanovesicles drawing increasing attention for their broad biosignaling functions, can shed new light on how multiple heterogeneous cells communicate for the purpose of bone remodeling. In the healthy bone, exosomes transmit signals favoring both bone synthesis and resorption, regulating the differentiation, recruitment, and activity of most cell types involved in bone remodeling and even assuming an active role in extracellular matrix mineralization. Additionally, in the ailing bone, they actively participate in pathogenic processes constituting also potential therapeutic agents and drug vectors. The present review summarizes the current knowledge on bone exosomes and bone remodeling in health and disease.
Collapse
|
202
|
Aglan HA, Ahmed HH, Mahmoud NS, Aly RM, Ali NA, Abd-Rabou AA. Nanotechnological Applications Hold a Pivotal Position in Boosting Stem Cells Osteogenic Activity: In Vitro and In Vivo Studies. Appl Biochem Biotechnol 2019; 190:551-573. [PMID: 31396888 DOI: 10.1007/s12010-019-03105-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/18/2019] [Indexed: 12/29/2022]
Abstract
This approach was constructed to appraise the therapeutic effectiveness of a single i.v. dose of osteoblasts generated from co-culturing BM-MSCs with nano-HA, Pt-NPs, or Pt-HA-nanocomposite in osteoporotic rats. MSCs were grown, propagated in culture, and characterized. The effect of the suggested nanoplatforms on the survival, osteogenic differentiation, and mineralization of BM-MSCs was assessed by MTT assay, real-time PCR analysis, and Alizarin red S staining, respectively. Thereafter, the generated osteoblasts were employed for the treatment of ovariectomized rats. Our results revealed that the selected nanoplatforms upregulate the expression of osteogenic differentiation related genes (Runx-2 and BMP-2) significantly and enhance calcium deposition in BM-MSCs after 7 and 21 days, respectively, whereas the in vivo study validated that the infusion of the generated osteoblasts considerably downturn serum BALP, BSP, and SOST levels; upswing OSX level; and regain femur bone mineralization and histoarchitecture. Conclusively, the outcomes of this work provide scientific evidence that transplanting osteoblasts derived from differentiation of BM-MSCs in the presence of nanoplatforms in ovariectomized rats restores bone remodeling balance which constitutes a new hope for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Hadeer A Aglan
- Hormones Department, Medical Research Division, National Research Centre, Giza, Egypt. .,Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt.
| | - Hanaa H Ahmed
- Hormones Department, Medical Research Division, National Research Centre, Giza, Egypt.,Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Nadia S Mahmoud
- Hormones Department, Medical Research Division, National Research Centre, Giza, Egypt.,Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Riham M Aly
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt.,Basic Dental Science Department, Oral & Dental Research Division, National Research Centre, Giza, Egypt
| | - Naglaa A Ali
- Hormones Department, Medical Research Division, National Research Centre, Giza, Egypt
| | - Ahmed A Abd-Rabou
- Hormones Department, Medical Research Division, National Research Centre, Giza, Egypt.,Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| |
Collapse
|
203
|
Mora-Boza A, López-Donaire ML, Saldaña L, Vilaboa N, Vázquez-Lasa B, San Román J. Glycerylphytate compounds with tunable ion affinity and osteogenic properties. Sci Rep 2019; 9:11491. [PMID: 31391524 PMCID: PMC6685941 DOI: 10.1038/s41598-019-48015-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/27/2019] [Indexed: 12/13/2022] Open
Abstract
Phytic acid (PA) is a natural-occurring antioxidant, which plays an important role in many biological processes. PA is recognized as a potent inhibitor of lipid peroxidation because of its high affinity to multivalent cations, and it can play a role in osteogenic processes. However, its powerful chelating capacity is controversial because it can lead to a severe reduction of mineral availability in the organism. For this reason, compounds with beneficial biological properties of PA, but a modular ion binding capacity, are of high interest. In this work, we report the synthesis and physicochemical characterization of two hydroxylic derivatives of PA, named glycerylphytates (GPhy), through a condensation reaction of PA with glycerol (G). Both derivatives present antioxidant properties, measured by ferrozine/FeCl2 method and chelating activity with calcium ions depending on the content of glyceryl groups incorporated. Besides, the hydroxylic modification not only modulates the ion binding affinity of derivatives but also improves their cytocompatibility in human bone marrow mesenchymal cells (MSCs). Furthermore, GPhy derivatives display osteogenic properties, confirmed by COL1A and ALPL expression depending on composition. These positive features convert GPhy compounds into potent alternatives for those skeletal diseases treatments where PA is tentatively applied.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Institute of Polymer Science and Technology, ICTP-CSIC, C/Juan de la Cierva 3, 28006, Madrid, Spain.,CIBER-BBN, Health Institute Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, 28029, Madrid, Spain
| | | | - Laura Saldaña
- CIBER-BBN, Health Institute Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, 28029, Madrid, Spain.,Hospital Universitario La Paz-IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
| | - Nuria Vilaboa
- Hospital Universitario La Paz-IdiPAZ, Paseo de La Castellana 261, 28046, Madrid, Spain
| | - Blanca Vázquez-Lasa
- Institute of Polymer Science and Technology, ICTP-CSIC, C/Juan de la Cierva 3, 28006, Madrid, Spain. .,CIBER-BBN, Health Institute Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, 28029, Madrid, Spain.
| | - Julio San Román
- Institute of Polymer Science and Technology, ICTP-CSIC, C/Juan de la Cierva 3, 28006, Madrid, Spain.,CIBER-BBN, Health Institute Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, 28029, Madrid, Spain
| |
Collapse
|
204
|
Steffi C, Shi Z, Kong CH, Chong SW, Wang D, Wang W. Use of Polyphenol Tannic Acid to Functionalize Titanium with Strontium for Enhancement of Osteoblast Differentiation and Reduction of Osteoclast Activity. Polymers (Basel) 2019; 11:E1256. [PMID: 31362449 PMCID: PMC6723407 DOI: 10.3390/polym11081256] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/18/2019] [Accepted: 07/25/2019] [Indexed: 12/22/2022] Open
Abstract
Implant anchorage remains a challenge, especially in porous osteoporotic bone with high osteoclast activity. The implant surface is modified with osteogenic molecules to stimulate osseointegration. Strontium (Sr) is known for its osteogenic and anti-osteoclastogenic effects. In this study, Sr was immobilized on a titanium (Ti) surface using bioinspired polyphenol tannic acid (pTAN) coating as an ad-layer (Ti-pTAN). Two separate coating techniques were employed for comparative analysis. In the first technique, Ti was coated with a tannic acid solution containing Sr (Ti-pTAN-1Stp). In the second method, Ti was first coated with pTAN, before being immersed in a SrCl2 solution to immobilize Sr on Ti-pTAN (Ti-pTAN-2Stp). Ti-pTAN-1Stp and Ti-pTAN-2Stp augmented the alkaline phosphatase activity, collagen secretion, osteocalcin production and calcium deposition of MC3T3-E1 cells as compared to those of Ti and Ti-pTAN. However, osteoclast differentiation of RAW 264.7, as studied by TRAP activity, total DNA, and multinucleated cell formation, were decreased on Ti-pTAN, Ti-pTAN-1Stp and Ti-pTAN-2Stp as compared to Ti. Of all the substrates, osteoclast activity on Ti-pTAN-2Stp was the lowest. Hence, an economical and simple coating technique using pTAN as an adlayer preserved the dual biological effects of Sr. These results indicate a promising new approach to tailoring the cellular responses of implant surfaces.
Collapse
Affiliation(s)
- Chris Steffi
- Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block Level 11, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Zhilong Shi
- Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block Level 11, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Chee Hoe Kong
- Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block Level 11, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Sue Wee Chong
- Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block Level 11, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Dong Wang
- Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block Level 11, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Wilson Wang
- Department of Orthopaedic Surgery, National University of Singapore, NUHS Tower Block Level 11, 1E Kent Ridge Road, Singapore 119228, Singapore.
| |
Collapse
|
205
|
Alvarez C, Monasterio G, Cavalla F, Córdova LA, Hernández M, Heymann D, Garlet GP, Sorsa T, Pärnänen P, Lee HM, Golub LM, Vernal R, Kantarci A. Osteoimmunology of Oral and Maxillofacial Diseases: Translational Applications Based on Biological Mechanisms. Front Immunol 2019; 10:1664. [PMID: 31379856 PMCID: PMC6657671 DOI: 10.3389/fimmu.2019.01664] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 07/03/2019] [Indexed: 12/23/2022] Open
Abstract
The maxillofacial skeleton is highly dynamic and requires a constant equilibrium between the bone resorption and bone formation. The field of osteoimmunology explores the interactions between bone metabolism and the immune response, providing a context to study the complex cellular and molecular networks involved in oro-maxillofacial osteolytic diseases. In this review, we present a framework for understanding the potential mechanisms underlying the immuno-pathobiology in etiologically-diverse diseases that affect the oral and maxillofacial region and share bone destruction as their common clinical outcome. These otherwise different pathologies share similar inflammatory pathways mediated by central cellular players, such as macrophages, T and B cells, that promote the differentiation and activation of osteoclasts, ineffective or insufficient bone apposition by osteoblasts, and the continuous production of osteoclastogenic signals by immune and local stromal cells. We also present the potential translational applications of this knowledge based on the biological mechanisms involved in the inflammation-induced bone destruction. Such applications can be the development of immune-based therapies that promote bone healing/regeneration, the identification of host-derived inflammatory/collagenolytic biomarkers as diagnostics tools, the assessment of links between oral and systemic diseases; and the characterization of genetic polymorphisms in immune or bone-related genes that will help diagnosis of susceptible individuals.
Collapse
Affiliation(s)
- Carla Alvarez
- Forsyth Institute, Cambridge, MA, United States
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Gustavo Monasterio
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Franco Cavalla
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Luis A. Córdova
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, San Jose's Hospital and Clínica Las Condes, Universidad de Chile, Santiago, Chile
| | - Marcela Hernández
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Dominique Heymann
- INSERM, UMR 1232, LabCT, CRCINA, Institut de Cancérologie de l'Ouest, Université de Nantes, Université d'Angers, Saint-Herblain, France
| | - Gustavo P. Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Department of Oral Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Pirjo Pärnänen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Hsi-Ming Lee
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Lorne M. Golub
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Dentistry Unit, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | | |
Collapse
|
206
|
Lerner UH, Kindstedt E, Lundberg P. The critical interplay between bone resorbing and bone forming cells. J Clin Periodontol 2019; 46 Suppl 21:33-51. [DOI: 10.1111/jcpe.13051] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/05/2018] [Accepted: 12/01/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Ulf H. Lerner
- Centre for Bone and Arthritis Research at Department of Internal Medicine and Clinical Nutrition; Institute of Medicine; Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
- Department of Odontology; Division of Molecular Periodontology; Umeå University; Umeå Sweden
| | - Elin Kindstedt
- Department of Odontology; Division of Molecular Periodontology; Umeå University; Umeå Sweden
| | - Pernilla Lundberg
- Department of Odontology; Division of Molecular Periodontology; Umeå University; Umeå Sweden
| |
Collapse
|
207
|
Wu N, Liu B, Du H, Zhao S, Li Y, Cheng X, Wang S, Lin J, Zhou J, Qiu G, Wu Z, Zhang J. The Progress of CRISPR/Cas9-Mediated Gene Editing in Generating Mouse/Zebrafish Models of Human Skeletal Diseases. Comput Struct Biotechnol J 2019; 17:954-962. [PMID: 31360334 PMCID: PMC6639410 DOI: 10.1016/j.csbj.2019.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/28/2019] [Accepted: 06/11/2019] [Indexed: 12/18/2022] Open
Abstract
Genetic factors play a substantial role in the etiology of skeletal diseases, which involve 1) defects in skeletal development, including intramembranous ossification and endochondral ossification; 2) defects in skeletal metabolism, including late bone growth and bone remodeling; 3) defects in early developmental processes related to skeletal diseases, such as neural crest cell (NCC) and cilia functions; 4) disturbance of the cellular signaling pathways which potentially affect bone growth. Efficient and high-throughput genetic methods have enabled the exploration and verification of disease-causing genes and variants. Animal models including mouse and zebrafish have been extensively used in functional mechanism studies of causal genes and variants. The conventional approaches of generating mutant animal models include spontaneous mutagenesis, random integration, and targeted integration via mouse embryonic stem cells. These approaches are costly and time-consuming. Recent development and application of gene-editing tools, especially the CRISPR/Cas9 system, has significantly accelerated the process of gene-editing in diverse organisms. Here we review both mice and zebrafish models of human skeletal diseases generated by CRISPR/Cas9 system, and their contributions to deciphering the underpins of disease mechanisms.
Collapse
Affiliation(s)
- Nan Wu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
- Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Bowen Liu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | - Huakang Du
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | - Sen Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | - Yaqi Li
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | - Xi Cheng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | - Shengru Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | - Jiachen Lin
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | - Junde Zhou
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
| | | | - Guixing Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
- Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing 100730, China
- Central Laboratory & Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Wu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
- Central Laboratory & Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguo Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing 100730, China
- Medical Research Center of Orthopedics, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
208
|
Park E, Kim J, Kim MC, Yeo S, Kim J, Park S, Jo M, Choi CW, Jin HS, Lee SW, Li WY, Lee JW, Park JH, Huh D, Jeong SY. Anti-Osteoporotic Effects of Kukoamine B Isolated from Lycii Radicis Cortex Extract on Osteoblast and Osteoclast Cells and Ovariectomized Osteoporosis Model Mice. Int J Mol Sci 2019; 20:ijms20112784. [PMID: 31174394 PMCID: PMC6600412 DOI: 10.3390/ijms20112784] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 06/06/2019] [Indexed: 11/16/2022] Open
Abstract
Osteoporosis is an abnormal bone remodeling condition characterized by decreased bone density, which leads to high risks of fracture. Previous study has demonstrated that Lycii Radicis Cortex (LRC) extract inhibits bone loss in ovariectomized (OVX) mice by enhancing osteoblast differentiation. A bioactive compound, kukoamine B (KB), was identified from fractionation of an LRC extract as a candidate component responsible for an anti-osteoporotic effect. This study investigated the anti-osteoporotic effects of KB using in vitro and in vivo osteoporosis models. KB treatment significantly increased the osteoblastic differentiation and mineralized nodule formation of osteoblastic MC3T3-E1 cells, while it significantly decreased the osteoclast differentiation of primary-cultured monocytes derived from mouse bone marrow. The effects of KB on osteoblastic and osteoclastic differentiations under more physiological conditions were also examined. In the co-culture of MC3T3-E1 cells and monocytes, KB promoted osteoblast differentiation but did not affect osteoclast differentiation. In vivo experiments revealed that KB significantly inhibited OVX-induced bone mineral density loss and restored the impaired bone structural properties in osteoporosis model mice. These results suggest that KB may be a potential therapeutic candidate for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Eunkuk Park
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Jeonghyun Kim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Mun-Chang Kim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Subin Yeo
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Jieun Kim
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Seulbi Park
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Miran Jo
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Chun Whan Choi
- Natural Products Research Institute, Gyeonggi Institute of Science & Technology Promotion, Suwon 16229, Korea.
| | - Hyun-Seok Jin
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan 31499, Korea.
| | - Sang Woo Lee
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea.
| | - Wan Yi Li
- Institute of Medicinal Plants, Yunnan Academy of Agricultural Sciences, Kunming 650200, China.
| | - Ji-Won Lee
- Korea Food Research Institute, Seongnam 13539, Korea.
| | - Jin-Hyok Park
- Dongwoodang Pharmacy Co. Ltd., Yeongchen 38819, Korea.
| | - Dam Huh
- Dongwoodang Pharmacy Co. Ltd., Yeongchen 38819, Korea.
| | - Seon-Yong Jeong
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| |
Collapse
|
209
|
Nehlin JO, Jafari A, Tencerova M, Kassem M. Aging and lineage allocation changes of bone marrow skeletal (stromal) stem cells. Bone 2019; 123:265-273. [PMID: 30946971 DOI: 10.1016/j.bone.2019.03.041] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/30/2019] [Accepted: 03/31/2019] [Indexed: 01/02/2023]
Abstract
Aging is associated with decreased bone mass and accumulation of bone marrow adipocytes. Both bone forming osteoblastic cells and bone marrow adipocytes are derived from a stem cell population within the bone marrow stroma called bone marrow stromal (skeletal or mesenchymal) stem cells (BMSC). In the present review, we provide an overview, based on the current literature, regarding the physiological aging processes that cause changes in BMSC lineage allocation, enhancement of adipocyte and defective osteoblast differentiation, leading to gradual exhaustion of stem cell regenerative potential and defects in bone tissue homeostasis and metabolism. We discuss strategies to preserve the "youthful" state of BMSC, to reduce bone marrow age-associated adiposity, and to counteract the overall negative effects of aging on bone tissues with the aim of decreasing bone fragility and risk of fractures.
Collapse
Affiliation(s)
- Jan O Nehlin
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Clinical Research Center, Copenhagen University Hospital, Hvidovre, Denmark.
| | - Abbas Jafari
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Department of Cellular and Molecular Medicine, The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Michaela Tencerova
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Danish Diabetes Academy, Novo Nordisk Foundation, Odense, Denmark
| | - Moustapha Kassem
- The Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark; Department of Cellular and Molecular Medicine, The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), Panum Institute, University of Copenhagen, Copenhagen, Denmark; Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
210
|
Chang Y, Cho B, Kim S, Kim J. Direct conversion of fibroblasts to osteoblasts as a novel strategy for bone regeneration in elderly individuals. Exp Mol Med 2019; 51:1-8. [PMID: 31073120 PMCID: PMC6509166 DOI: 10.1038/s12276-019-0251-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/24/2018] [Accepted: 01/28/2019] [Indexed: 12/31/2022] Open
Abstract
Mortality caused by age-related bone fractures or osteoporosis is steadily increasing worldwide as the population ages. The pace of the development of bone regeneration engineering to treat bone fractures has consequently increased in recent years. A range of techniques for bone regeneration, such as immunotherapy, allografts, and hydrogel therapy, have been devised. Cell-based therapies using bone marrow-derived mesenchymal stem cells and induced pluripotent stem cells derived from somatic cells are considered to be suitable approaches for bone repair. However, these cell-based therapies suffer from a number of limitations in terms of efficiency and safety. Somatic cells can also be directly differentiated into osteoblasts by several transcription factors. As osteoblasts play a central role in the process of bone formation, the direct reprogramming of fibroblasts into osteoblasts may hence be a new way to treat bone fractures in elderly individuals. Here, we review recent developments regarding the therapeutic potential of the direct reprogramming of cells for bone regeneration. Reprogramming cells that produce connective tissue to form bone instead could help prevent fractures in the elderly. Bones weaken with age, and fractures are a significant health risk in ageing populations. Most current bone regeneration treatments use stem cells, which can differentiate into any type of cell and have infinite capacity to divide; however, they are difficult to source and can lead to tumor formation. Jongpil Kim at Dongguk University in South Korea and coworkers have reviewed a new method that uses genetic signals to transform connective tissue-forming cells into bone-producing cells. The reprogrammed cells have been shown to generate new bone at the desired site, and because they have already lost their capacity for infinite division, tumor formation risk is greatly reduced. This method shows promise to expand treatment options for fractures and osteoporosis.
Collapse
Affiliation(s)
- Yujung Chang
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Byounggook Cho
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Siyoung Kim
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea
| | - Jongpil Kim
- Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea. .,Department of Chemistry, Dongguk University, 30, Pildong-ro 1-gil 30, Jung-gu, Seoul, 04620, Republic of Korea.
| |
Collapse
|
211
|
Shiraki M, Xu X, Iovanna JL, Kukita T, Hirata H, Kamohara A, Kubota Y, Miyamoto H, Mawatari M, Kukita A. Deficiency of stress-associated gene Nupr1 increases bone volume by attenuating differentiation of osteoclasts and enhancing differentiation of osteoblasts. FASEB J 2019; 33:8836-8852. [PMID: 31067083 DOI: 10.1096/fj.201802322rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nuclear protein 1 (NUPR1) is a multifunctional stress-induced protein involved in regulating tumorigenesis, apoptosis, and autophagy. Bone homeostasis is maintained by bone-resorbing osteoclasts and bone-forming osteoblasts and osteocytes. We aimed to determine the role of NUPR1 in bone metabolism. Using microcomputed tomography, we found that mice lacking Nupr1 exhibited increased bone volume. Histologic analysis showed that Nupr1 deficiency decreased osteoclast numbers but increased osteoblast numbers and osteoid formation. In vitro culture of bone marrow macrophages showed that receptor activator of NF-κB ligand-induced osteoclastogenesis was down-regulated in Nupr1-deficient mice. In contrast, primary osteoblasts from Nupr1-deficient mice revealed that proliferation of osteoblasts and expression of bone matrix proteins were markedly enhanced. In addition, expression of autophagy-related genes, formation of autophagosomes, and cell survival were up-regulated in Nupr1-deficient osteoblasts. In contract, deletion of Nupr1 reduced the formation of osteocyte cellular projection, which is an indicator of mature osteocytes. Importantly, we found that the expression of sclerostin (Sost), an inhibitor of bone formation, was down-regulated in the osteoblasts and osteocytes of Nupr1-deficient mice. Conversely, Nupr1 overexpression enhanced Sost expression in primary osteoblasts. Collectively, these results indicate that Nupr1 deficiency increases bone volume by attenuating production of Sost and osteoclastogenesis and enhancing differentiation of osteoblasts.-Shiraki, M., Xu, X., Iovanna, J. L., Kukita, T., Hirata, H., Kamohara, A., Kubota, Y., Miyamoto, H., Mawatari, M., Kukita, A. Deficiency of stress-associated gene Nupr1 increases bone volume by attenuating differentiation of osteoclasts and enhancing differentiation of osteoblasts.
Collapse
Affiliation(s)
- Makoto Shiraki
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Xianghe Xu
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.,Department of Molecular Cell Biology and Oral Anatomy, Kyushu University, Fukuoka, Japan
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U 1068, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France; and
| | - Toshio Kukita
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University, Fukuoka, Japan
| | - Hirohito Hirata
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Asana Kamohara
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Yasushi Kubota
- Division of Hematology, Respiratory Medicine, and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hiroshi Miyamoto
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Masaaki Mawatari
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Akiko Kukita
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
212
|
Systemic administration of low-dose naltrexone increases bone mass due to blockade of opioid growth factor receptor signaling in mice osteoblasts. Life Sci 2019; 224:232-240. [DOI: 10.1016/j.lfs.2019.03.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/14/2019] [Accepted: 03/27/2019] [Indexed: 01/31/2023]
|
213
|
Bighetti ACC, Cestari TM, Santos PS, Arantes RVN, Paini S, Assis GF, Costa BC, de Oliveira FA, Tokuhara CK, de Oliveira RC, Taga R. In vitro and in vivo assessment of CaP materials for bone regenerative therapy. The role of multinucleated giant cells/osteoclasts in bone regeneration. J Biomed Mater Res B Appl Biomater 2019; 108:282-297. [PMID: 31009176 DOI: 10.1002/jbm.b.34388] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/22/2019] [Accepted: 03/30/2019] [Indexed: 12/15/2022]
Abstract
In this work, bone formation/remodeling/maturation was correlated with the presence of multinucleated giant cells (MGCs)/osteoclasts (tartrate-resistant acid phosphatase [TRAP]-positive cells) on the surface of beta-tricalcium phosphate (β-TCP), sintered deproteinized bovine bone (sDBB), and carbonated deproteinized bovine bone (cDBB) using a maxillary sinus augmentation (MSA) in a New Zealand rabbit model. Microtomographic, histomorphometric, and immunolabeling for TRAP-cells analyses were made at 15, 30, and 60 days after surgery. In all treatments, a faster bone formation/remodeling/maturation and TRAP-positive cells activity occurred in the osteotomy region of the MSA than in the middle and submucosa regions. In the β-TCP, the granules were rapidly reabsorbed by TRAP-positive cells and replaced by bone tissue. β-TCP enabled quick bone regeneration/remodeling and full bone and marrow restoration until 60 days, but with a significant reduction in MSA volume. In cDBB and sDBB, the quantity of TRAP-positive cells was smaller than in β-TCP, and these cells were associated with granule surface preparation for osteoblast-mediated bone formation. After 30 days, more than 80% of granule surfaces were surrounded and integrated by bone tissue without signs of degradation, preserving the MSA volume. Overall, the materials tested in a standardized preclinical model led to different bone formation/remodeling/maturation within the same repair process influenced by different microenvironments and MGCs/osteoclasts. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 108B:282-297, 2020.
Collapse
Affiliation(s)
- Ana Carolina Cestari Bighetti
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Tania Mary Cestari
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Paula Sanches Santos
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Ricardo Vinicius Nunes Arantes
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Suelen Paini
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Gerson Francisco Assis
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Bruna Carolina Costa
- Physics Department, Advanced Materials Laboratory, São Paulo State University, UNESP, Avenue Luiz Edmundo Carrijo Coube 14-01, Bauru, São Paulo, 17033-360, Brazil
| | - Flávia Amadeu de Oliveira
- Laboratory of Biochemistry of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Cintia Kazuko Tokuhara
- Laboratory of Biochemistry of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Rodrigo Cardoso de Oliveira
- Laboratory of Biochemistry of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| | - Rumio Taga
- Laboratory of Histology of Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, 17012-901, Brazil
| |
Collapse
|
214
|
Liu X, Wan M. A tale of the good and bad: Cell senescence in bone homeostasis and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:97-128. [PMID: 31122396 DOI: 10.1016/bs.ircmb.2019.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Historically, cellular senescence has been viewed as an irreversible cell-cycle arrest process with distinctive phenotypic alterations that were implicated primarily in aging and tumor suppression. Recent discoveries suggest that cellular senescence represents a series of diverse, dynamic, and heterogeneous cellular states with the senescence-associated secretory phenotype (SASP). Although senescent cells typically contribute to aging and age-related diseases, accumulating evidence has shown that they also have important physiological functions during embryonic development, late pubertal bone growth cessation, and adulthood tissue remodeling. Here, we review the recent research on cellular senescence and SASP, highlighting the key pathways that mediate senescence cell-cycle arrest and initiate SASP. We also summarize recent literature on the role of cellular senescence in maintaining bone homeostasis and mediating age-associated osteoporosis, discussing both the beneficial and adverse roles of cellular senescence in bone during different physiological stages, including bone development, childhood bone growth, adulthood bone remodeling, and bone aging.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
215
|
Humbert P, Brennan MÁ, Davison N, Rosset P, Trichet V, Blanchard F, Layrolle P. Immune Modulation by Transplanted Calcium Phosphate Biomaterials and Human Mesenchymal Stromal Cells in Bone Regeneration. Front Immunol 2019; 10:663. [PMID: 31001270 PMCID: PMC6455214 DOI: 10.3389/fimmu.2019.00663] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/11/2019] [Indexed: 12/22/2022] Open
Abstract
A wide variety of biomaterials have been developed as both stabilizing structures for the injured bone and inducers of bone neoformation. They differ in chemical composition, shape, porosity, and mechanical properties. The most extensively employed and studied subset of bioceramics are calcium phosphate materials (CaPs). These materials, when transplanted alongside mesenchymal stem cells (MSCs), lead to ectopic (intramuscular and subcutaneous) and orthotopic bone formation in preclinical studies, and effective fracture healing in clinical trials. Human MSC transplantation in pre-clinical and clinical trials reveals very low engraftment in spite of successful clinical outcomes and their therapeutic actions are thought to be primarily through paracrine mechanisms. The beneficial role of transplanted MSC could rely on their strong immunomodulatory effect since, even without long-term engraftment, they have the ability to alter both the innate and adaptive immune response which is critical to facilitate new bone formation. This study presents the current knowledge of the immune response to the implantation of CaP biomaterials alone or in combination with MSC. In particular the central role of monocyte-derived cells, both macrophages and osteoclasts, in MSC-CaP mediated bone formation is emphasized. Biomaterial properties, such as macroporosity and surface microstructure, dictate the host response, and the ultimate bone healing cascade. Understanding intercellular communications throughout the inflammation, its resolution and the bone regeneration phase, is crucial to improve the current therapeutic strategies or develop new approaches.
Collapse
Affiliation(s)
- Paul Humbert
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | - Meadhbh Á. Brennan
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Harvard School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Noel Davison
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- Instructure Labs, B.V., The Hague, Netherlands
| | - Philippe Rosset
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
- Centre Hospitalier Universitaire de Tours, Tours, France
| | - Valérie Trichet
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| | | | - Pierre Layrolle
- Laboratory Phy-Os, Inserm UMR1238, University of Nantes, Nantes, France
| |
Collapse
|
216
|
Park SSH, Zhang L, Attia T, Salat P, Banks K, Willett T, Grynpas M. Pre-clinical evaluation of bone allograft toughened with a novel sterilization method: An in vivo rabbit study. J Orthop Res 2019; 37:832-844. [PMID: 30839120 DOI: 10.1002/jor.24269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 02/12/2019] [Indexed: 02/04/2023]
Abstract
Bone allografts often undergo γ-irradiation sterilization to decrease infection risk. However this consequently degrades bone collagen and makes the allograft brittle. Our laboratory has previously found that pre-treatment with ribose ex vivo protects the bone. However, it remains unclear whether or not ribose-treated γ-irradiated allografts are able to unite and remodel in vivo. Using New Zealand White rabbits (NZWr), we aimed to evaluate if ribose-treated allografts can unite with host bone (compared to untreated (fresh-frozen) and conventionally-irradiated allografts). A critically-sized defect was created in the radii of NZWr and reconstructed with allografts fixed with an intramedullary Kirschner wire. Healing and union were assessed at 2, 6, and 12 weeks post operation, with radiographs, µCT, static and dynamic histomorphometry, backscatter electron microscopy, and torsion testing. Intramedullary fixation achieved stable reconstructions and bony union in all groups and no differences were found in the radiographic and biomechanical parameters tested. Interestingly, γ-irradiated allografts had significantly less bone volume due to evident resorption of the grafts. In contrast, ribose pre-treatment protected γ-irradiated allografts from this bone loss, with results similar to the fresh frozen controls. In conclusion, ribose-pretreated γ-irradiated allografts were able to unite in vivo. In addition to achieving bony union with host bone, ribose pre-treatment may protect against allograft resorption. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Sam Si-Hyeong Park
- Department of Surgery, Division of Orthopaedic Surgery, University of Toronto, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Lucia Zhang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Tarik Attia
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Peter Salat
- Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Kate Banks
- Division of Comparative Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Thomas Willett
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Marc Grynpas
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| |
Collapse
|
217
|
Li Z, Liu T, Gilmore A, Gómez NM, Mitchell CH, Li YP, Oursler MJ, Yang S. Regulator of G Protein Signaling Protein 12 (Rgs12) Controls Mouse Osteoblast Differentiation via Calcium Channel/Oscillation and Gαi-ERK Signaling. J Bone Miner Res 2019; 34:752-764. [PMID: 30489658 PMCID: PMC7675783 DOI: 10.1002/jbmr.3645] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/13/2018] [Accepted: 11/17/2018] [Indexed: 12/11/2022]
Abstract
Bone homeostasis intimately relies on the balance between osteoblasts (OBs) and osteoclasts (OCs). Our previous studies have revealed that regulator of G protein signaling protein 12 (Rgs12), the largest protein in the Rgs super family, is essential for osteoclastogenesis from hematopoietic cells and OC precursors. However, how Rgs12 regulates OB differentiation and function is still unknown. To understand that, we generated an OB-targeted Rgs12 conditional knockout (CKO) mice model by crossing Rgs12fl/fl mice with Osterix (Osx)-Cre transgenic mice. We found that Rgs12 was highly expressed in both OB precursor cells (OPCs) and OBs of wild-type (WT) mice, and gradually increased during OB differentiation, whereas Rgs12-CKO mice (OsxCre/+ ; Rgs12fl/fl ) exhibited a dramatic decrease in both trabecular and cortical bone mass, with reduced numbers of OBs and increased apoptotic cell population. Loss of Rgs12 in OPCs in vitro significantly inhibited OB differentiation and the expression of OB marker genes, resulting in suppression of OB maturation and mineralization. Further mechanism study showed that deletion of Rgs12 in OPCs significantly inhibited guanosine triphosphatase (GTPase) activity and cyclic adenosine monophosphate (cAMP) level, and impaired Calcium (Ca2+ ) oscillations via restraints of major Ca2+ entry sources (extracellular Ca2+ influx and intracellular Ca2+ release from endoplasmic reticulum), partially contributed by the blockage of L-type Ca2+ channel mediated Ca2+ influx. Downstream mediator extracellular signal-related protein kinase (ERK) was found inactive in OBs of OsxCre/+ ; Rgs12fl/fl mice and in OPCs after Rgs12 deletion, whereas application of pertussis toxin (PTX) or overexpression of Rgs12 could rescue the defective OB differentiation via restoration of ERK phosphorylation. Our findings reveal that Rgs12 is an important regulator during osteogenesis and highlight Rgs12 as a potential therapeutic target for bone disorders. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ziqing Li
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Tongjun Liu
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
- Department of Implantology, Shandong Provincial Key Laboratory of Oral Biomedicine, School of Stomatology, Shandong University
- Department of Stomatology, the Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong province 250000, China
| | - Alyssa Gilmore
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
| | - Néstor Más Gómez
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- Department of Physiology, School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Yi-ping Li
- Department of Pathology, University of Alabama in Birmingham, Birmingham, AL 35294, USA
| | - Merry J Oursler
- Department of Medicine, Endocrine Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Shuying Yang
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- The Penn Center for Musculoskeletal Disorders, University of Pennsylvania Philadelphia, PA 19104, USA
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
| |
Collapse
|
218
|
Kim BJ, Koh JM. Coupling factors involved in preserving bone balance. Cell Mol Life Sci 2019; 76:1243-1253. [PMID: 30515522 PMCID: PMC11105749 DOI: 10.1007/s00018-018-2981-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/14/2018] [Accepted: 11/26/2018] [Indexed: 12/11/2022]
Abstract
Coupling during bone remodeling refers to the spatial and temporal coordination of bone resorption with bone formation. Studies have assessed the subtle interactions between osteoclasts and osteoblasts to preserve bone balance. Traditionally, coupling research related to osteoclast function has focused on bone resorption activity causing the release of growth factors embedded in the bone matrix. However, considerable evidence from in vitro, animal, and human studies indicates the importance of the osteoclasts themselves in coupling phenomena, and many osteoclast-derived coupling factors have been identified. These include sphingosine-1-phosphate, vesicular-receptor activator of nuclear factor-κB, collagen triple helix repeat containing 1, and cardiotrophin-1. Interestingly, neuronal guidance molecules, such as slit guidance ligand 3, semaphorin (SEMA) 3A, SEMA4D, and netrin-1, originally identified as instructive cues allowing the navigation of growing axons to their targets, have been shown to be involved in the intercellular cross-talk among bone cells. This review discusses osteoclast-osteoblast coupling signals, including recent advances and the potential roles of these signals as therapeutic targets for osteoporosis and as biomarkers predicting human bone health.
Collapse
Affiliation(s)
- Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
219
|
Hasegawa T, Kikuta J, Ishii M. Imaging the Bone-Immune Cell Interaction in Bone Destruction. Front Immunol 2019; 10:596. [PMID: 30972080 PMCID: PMC6443987 DOI: 10.3389/fimmu.2019.00596] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/05/2019] [Indexed: 11/13/2022] Open
Abstract
Bone is a highly dynamic organ that is continuously being remodeled by the reciprocal interactions between bone and immune cells. We have originally established an advanced imaging system for visualizing the in vivo behavior of osteoclasts and their precursors in the bone marrow cavity using two-photon microscopy. Using this system, we found that the blood-enriched lipid mediator, sphingosine-1-phosphate, controlled the migratory behavior of osteoclast precursors. We also developed pH-sensing chemical fluorescent probes to detect localized acidification by bone-resorbing osteoclasts on the bone surface in vivo, and identified two distinct functional states of differentiated osteoclasts, "bone-resorptive" and "non-resorptive." Here, we summarize our studies on the dynamics and functions of bone and immune cells within the bone marrow. We further discuss how our intravital imaging techniques can be applied to evaluate the mechanisms of action of biological agents in inflammatory bone destruction. Our intravital imaging techniques would be beneficial for studying the cellular dynamics in arthritic inflammation and bone destruction in vivo and would also be useful for evaluating novel therapies in animal models of bone-destroying diseases.
Collapse
Affiliation(s)
- Tetsuo Hasegawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan.,Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan.,WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan.,WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
220
|
Saito H, Gasser A, Bolamperti S, Maeda M, Matthies L, Jähn K, Long CL, Schlüter H, Kwiatkowski M, Saini V, Pajevic PD, Bellido T, van Wijnen AJ, Mohammad KS, Guise TA, Taipaleenmäki H, Hesse E. TG-interacting factor 1 (Tgif1)-deficiency attenuates bone remodeling and blunts the anabolic response to parathyroid hormone. Nat Commun 2019; 10:1354. [PMID: 30902975 PMCID: PMC6430773 DOI: 10.1038/s41467-019-08778-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 01/29/2019] [Indexed: 01/29/2023] Open
Abstract
Osteoporosis is caused by increased bone resorption and decreased bone formation. Intermittent administration of a fragment of Parathyroid hormone (PTH) activates osteoblast-mediated bone formation and is used in patients with severe osteoporosis. However, the mechanisms by which PTH elicits its anabolic effect are not fully elucidated. Here we show that the absence of the homeodomain protein TG-interacting factor 1 (Tgif1) impairs osteoblast differentiation and activity, leading to a reduced bone formation. Deletion of Tgif1 in osteoblasts and osteocytes decreases bone resorption due to an increased secretion of Semaphorin 3E (Sema3E), an osteoclast-inhibiting factor. Tgif1 is a PTH target gene and PTH treatment failed to increase bone formation and bone mass in Tgif1-deficient mice. Thus, our study identifies Tgif1 as a novel regulator of bone remodeling and an essential component of the PTH anabolic action. These insights contribute to a better understanding of bone metabolism and the anabolic function of PTH.
Collapse
Affiliation(s)
- Hiroaki Saito
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Andreas Gasser
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Simona Bolamperti
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Miki Maeda
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Levi Matthies
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Katharina Jähn
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Courtney L Long
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Hartmut Schlüter
- Mass Spectrometric Proteomics Laboratory, Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Marcel Kwiatkowski
- Mass Spectrometric Proteomics Laboratory, Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Vaibhav Saini
- Endocrine Unit, Massachusetts General Hospital, 55 Fruit St., Boston, MA, 02114, USA
| | - Paola Divieti Pajevic
- Endocrine Unit, Massachusetts General Hospital, 55 Fruit St., Boston, MA, 02114, USA
- Department of Molecular and Cell Biology, Boston University, School of Dental Medicine, 72 East Concord St., Boston, MA, 02118, USA
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Khalid S Mohammad
- Division of Endocrinology, Department of Medicine, Indiana School of Medicine, 545 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Theresa A Guise
- Division of Endocrinology, Department of Medicine, Indiana School of Medicine, 545 Barnhill Dr., Indianapolis, IN, 46202, USA
| | - Hanna Taipaleenmäki
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - Eric Hesse
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany.
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., Indianapolis, IN, 46202, USA.
| |
Collapse
|
221
|
Eick GN, Devlin MJ, Cepon‐Robins TJ, Kowal P, Sugiyama LS, Snodgrass JJ. A dried blood spot‐based method to measure levels of tartrate‐resistant acid phosphatase 5b (TRACP‐5b), a marker of bone resorption. Am J Hum Biol 2019; 31:e23240. [DOI: 10.1002/ajhb.23240] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 02/08/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
Affiliation(s)
- Geeta N. Eick
- Department of Anthropology University of Oregon Eugene Oregon
| | - Maureen J. Devlin
- Department of Anthropology University of Michigan Ann Arbor Michigan
| | | | - Paul Kowal
- Department of Anthropology University of Oregon Eugene Oregon
- Department of Health Statistics and Information Systems World Health Organization Genève Switzerland
- University of Newcastle Research Centre for Generational Health and Ageing Newcastle New South Wales Australia
| | | | | |
Collapse
|
222
|
Nakano S, Inoue K, Xu C, Deng Z, Syrovatkina V, Vitone G, Zhao L, Huang XY, Zhao B. G-protein Gα 13 functions as a cytoskeletal and mitochondrial regulator to restrain osteoclast function. Sci Rep 2019; 9:4236. [PMID: 30862896 PMCID: PMC6414604 DOI: 10.1038/s41598-019-40974-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/19/2019] [Indexed: 12/13/2022] Open
Abstract
Excessive osteoclastic bone erosion disrupts normal bone remodeling and leads to bone loss in many skeletal diseases, including inflammatory arthritis, such as rheumatoid arthritis (RA) and psoriatic arthritis, periodontitis and peri-prosthetic loosening. Functional control of osteoclasts is critical for the maintenance of bone homeostasis. However, the mechanisms that restrain osteoclast resorptive function are not fully understood. In this study, we identify a previously unrecognized role for G-protein Gα13 in inhibition of osteoclast adhesion, fusion and bone resorptive function. Gα13 is highly expressed in mature multinucleated osteoclasts, but not during early differentiation. Deficiency of Gα13 in myeloid osteoclast lineage (Gα13ΔM/ΔM mice) leads to super spread morphology of multinucleated giant osteoclasts with elevated bone resorptive capacity, corroborated with an osteoporotic bone phenotype in the Gα13ΔM/ΔM mice. Mechanistically, Gα13 functions as a brake that restrains the c-Src, Pyk2, RhoA-Rock2 mediated signaling pathways and related gene expressions to control the ability of osteoclasts in fusion, adhesion, actin cytoskeletal remodeling and resorption. Genome wide analysis reveals cytoskeleton related genes that are suppressed by Gα13, identifying Gα13 as a critical cytoskeletal regulator in osteoclasts. We also identify a genome wide regulation of genes responsible for mitochondrial biogenesis and function by Gα13 in osteoclasts. Furthermore, the significant correlation between Gα13 expression levels, TNF activity and RA disease activity in RA patients suggests that the Gα13 mediated mechanisms represent attractive therapeutic targets for diseases associated with excessive bone resorption.
Collapse
Affiliation(s)
- Shinichi Nakano
- Arthritis and Tissue Degeneration Program and The David Z, Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Kazuki Inoue
- Arthritis and Tissue Degeneration Program and The David Z, Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Cheng Xu
- Arthritis and Tissue Degeneration Program and The David Z, Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Zhonghao Deng
- Arthritis and Tissue Degeneration Program and The David Z, Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Viktoriya Syrovatkina
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Gregory Vitone
- Arthritis and Tissue Degeneration Program and The David Z, Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Liang Zhao
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and The David Z, Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA.
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA.
- Graduate Program in Cell & Developmental Biology, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA.
| |
Collapse
|
223
|
Shiwaku Y, Tsuchiya K, Xiao L, Suzuki O. Effect of calcium phosphate phases affecting the crosstalk between osteoblasts and osteoclasts in vitro. J Biomed Mater Res A 2019; 107:1001-1013. [PMID: 30684383 DOI: 10.1002/jbm.a.36626] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/12/2018] [Indexed: 11/08/2022]
Abstract
Previous studies have reported that octacalcium phosphate (OCP) enhances osteoblast differentiation and osteoclast formation during the hydrolysis process to hydroxyapatite (HA). However, the crystal phases that affect the crosstalk between osteoclasts and osteoblasts are unknown, which should determine the bone substitute material's property of OCP. The present study was designed to investigate whether the chemical composition and crystal structure of calcium phosphates affect osteoclast formation and the osteoclast-osteoblast crosstalk. Biodegradable β-tricalcium phosphate (β-TCP) was used as the control material. Osteoclasts were cultured on HA/OCP or HA/TCP disks and their cellular responses were assessed. Both OCP and β-TCP had a similar ability to create multinucleated osteoclasts. However, OCP promoted the expression of complement component 3a (C3a), a positive coupling factor, in osteoclasts, whereas β-TCP enhanced that of EphrinB2 (EfnB2) and collagen triple helix repeat containing 1 (Cthrc1). During osteoclast culture, phosphate ions were released from the crystals, and OCP-HA conversion was advanced in HA/OCP mixtures and OCP. X-ray diffraction analysis revealed no remarkable changes in the crystal structures of HA/TCP mixtures and β-TCP before and after osteoclast culture. These results indicate that the distinct chemical environment induced by the calcium phosphate phases affects the crosstalk between osteoclasts and osteoblasts. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1001-1013, 2019.
Collapse
Affiliation(s)
- Yukari Shiwaku
- Division of Craniofacial Function Engineering, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kaori Tsuchiya
- Division of Craniofacial Function Engineering, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Linghao Xiao
- Division of Craniofacial Function Engineering, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Division of Advanced Prosthetic Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Osamu Suzuki
- Division of Craniofacial Function Engineering, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
224
|
Chen L, Shi K, Andersen TL, Qiu W, Kassem M. KIAA1199 is a secreted molecule that enhances osteoblastic stem cell migration and recruitment. Cell Death Dis 2019; 10:126. [PMID: 30755597 PMCID: PMC6372631 DOI: 10.1038/s41419-018-1202-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022]
Abstract
Factors mediating mobilization of osteoblastic stem and progenitor cells from their bone marrow niche to be recruited to bone formation sites during bone remodeling are poorly known. We have studied secreted factors present in the bone marrow microenvironment and identified KIAA1199 (also known as CEMIP, cell migration inducing hyaluronan binding protein) in human bone biopsies as highly expressed in osteoprogenitor reversal cells (Rv.C) recruited to the eroded surfaces (ES), which are the future bone formation sites. In vitro, KIAA1199 did not affect the proliferation of human osteoblastic stem cells (also known as human bone marrow skeletal or stromal stem cells, hMSCs); but it enhanced cell migration as determined by scratch assay and trans-well migration assay. KIAA1199 deficient hMSCs (KIAA1199down) exhibited significant changes in cell size, cell length, ratio of cell width to length and cell roundness, together with reduction of polymerization actin (F-actin) and changes in phos-CFL1 (cofflin1), phos-LIMK1 (LIM domain kinase 1) and DSTN (destrin), key factors regulating actin cytoskeletal dynamics and cell motility. Moreover, KIAA1199down hMSC exhibited impaired Wnt signaling in TCF-reporter assay and decreased expression of Wnt target genes and these effects were rescued by KIAA1199 treatment. Finally, KIAA1199 regulated the activation of P38 kinase and its associated changes in Wnt-signaling. Thus, KIAA1199 is a mobilizing factor that interacts with P38 and Wnt signaling, and induces changes in actin cytoskeleton, as a mechanism mediating recruitment of hMSC to bone formation sites.
Collapse
Affiliation(s)
- Li Chen
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
| | - Kaikai Shi
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology, Vejle Hospital-Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, 7100, Vejle, Denmark
| | - Weimin Qiu
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital and University of Southern Denmark, 5000, Odense, Denmark.
- The Danish Stem Cell Center (DanStem), University of Copenhagen, 2200, Copenhagen, Denmark.
- Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
225
|
Myelofibrosis osteoclasts are clonal and functionally impaired. Blood 2019; 133:2320-2324. [PMID: 30745304 DOI: 10.1182/blood-2018-10-878926] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
Bone marrow (BM) sclerosis is commonly found in patients with late-stage myelofibrosis (MF). Because osteoclasts (OCs) and osteoblasts play a key role in bone remodeling, and MF monocytes, the OC precursors, are derived from the neoplastic clone, we wondered whether decreased OC numbers or impairment in their osteolytic function affects the development of osteosclerosis. Analysis of BM biopsies from 50 MF patients showed increased numbers of multinucleated tartrate-resistant acid phosphatase (TRAP)/cathepsin K+ OCs expressing phosphorylated Janus kinase 2 (JAK2). Randomly microdissected TRAP+ OCs from 16 MF patients harbored JAK2 or calreticulin (CALR) mutations, confirming MF OCs are clonal. To study OC function, CD14+ monocytes from MF patients and healthy individuals were cultured and differentiated into OCs. Unlike normal OCs, MF OCs appeared small and round, with few protrusions, and carried the mutations and chromosomal abnormalities of neoplastic clones. In addition, MF OCs lacked F-actin-rich ring-like structures and had fewer nuclei and reduced colocalization signals, compatible with decreased fusion events, and their mineral resorption capacity was significantly reduced, indicating impaired osteolytic function. Taken together, our data suggest that, although the numbers of MF OCs are increased, their impaired osteolytic activity distorts bone remodeling and contributes to the induction of osteosclerosis.
Collapse
|
226
|
Zimmerman SM, Dimori M, Heard-Lipsmeyer ME, Morello R. The Osteocyte Transcriptome Is Extensively Dysregulated in Mouse Models of Osteogenesis Imperfecta. JBMR Plus 2019; 3:e10171. [PMID: 31372585 PMCID: PMC6659450 DOI: 10.1002/jbm4.10171] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Osteocytes are long‐lived, highly interconnected, terminally differentiated osteoblasts that reside within mineralized bone matrix. They constitute about 95% of adult bone cells and play important functions including in the regulation of bone remodeling, phosphate homeostasis, and mechanical stimuli sensing and response. However, the role of osteocytes in the pathogenesis of congenital diseases of abnormal bone matrix is poorly understood. This study characterized in vivo transcriptional changes in osteocytes from CrtapKO and oim/oim mouse models of osteogenesis imperfecta (OI) compared with wild‐type (WT) control mice. To do this, RNA was extracted from osteocyte‐enriched cortical femurs and tibias, sequenced and subsequently analyzed to identify differentially expressed transcripts. These models were chosen because they mimic two types of OI with different genetic mutations that result in distinct type I collagen defects. A large number of transcripts were dysregulated in either model of OI, but 281 of them were similarly up‐ or downregulated in both compared with WT controls. Conversely, very few transcripts were differentially expressed between the CrtapKO and oim/oim mice, indicating that distinct alterations in type I collagen can lead to shared pathogenic processes and similar phenotypic outcomes. Bioinformatics analyses identified several critical hubs of dysregulation that were enriched in annotation terms such as development and differentiation, ECM and collagen fibril organization, cell adhesion, signaling, regulatory processes, pattern binding, chemotaxis, and cell projections. The data further indicated alterations in important signaling pathways such as WNT and TGF‐β but also highlighted new candidate genes to pursue in future studies. Overall, our study suggested that the osteocyte transcriptome is broadly dysregulated in OI with potential long‐term consequences at the cellular level, which deserve further investigations. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sarah M Zimmerman
- Department of Physiology and Biophysics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Milena Dimori
- Department of Physiology and Biophysics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Melissa E Heard-Lipsmeyer
- Department of Physiology and Biophysics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Roy Morello
- Department of Physiology and Biophysics University of Arkansas for Medical Sciences Little Rock AR USA.,Department of Orthopaedic Surgery University of Arkansas for Medical Sciences Little Rock AR USA.,Division of Genetics University of Arkansas for Medical Sciences Little Rock AR USA
| |
Collapse
|
227
|
Wasilewski GB, Vervloet MG, Schurgers LJ. The Bone-Vasculature Axis: Calcium Supplementation and the Role of Vitamin K. Front Cardiovasc Med 2019; 6:6. [PMID: 30805347 PMCID: PMC6370658 DOI: 10.3389/fcvm.2019.00006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Calcium supplements are broadly prescribed to treat osteoporosis either as monotherapy or together with vitamin D to enhance calcium absorption. It is still unclear whether calcium supplementation significantly contributes to the reduction of bone fragility and fracture risk. Data suggest that supplementing post-menopausal women with high doses of calcium has a detrimental impact on cardiovascular morbidity and mortality. Chronic kidney disease (CKD) patients are prone to vascular calcification in part due to impaired phosphate excretion. Calcium-based phosphate binders further increase risk of vascular calcification progression. In both bone and vascular tissue, vitamin K-dependent processes play an important role in calcium homeostasis and it is tempting to speculate that vitamin K supplementation might protect from the potentially untoward effects of calcium supplementation. This review provides an update on current literature on calcium supplementation among post-menopausal women and CKD patients and discusses underlying molecular mechanisms of vascular calcification. We propose therapeutic strategies with vitamin K2 treatment to prevent or hold progression of vascular calcification as a consequence of excessive calcium intake.
Collapse
Affiliation(s)
- Grzegorz B Wasilewski
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands.,Nattopharma ASA, Hovik, Norway
| | - Marc G Vervloet
- Department of Nephrology and Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
228
|
Identification of substrate-specific inhibitors of cathepsin K through high-throughput screening. Biochem J 2019; 476:499-512. [DOI: 10.1042/bcj20180851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/27/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Abstract
Cathepsin K (CatK) is a cysteine protease and drug target for skeletal disorders that is known for its potent collagenase and elastase activity. The formation of oligomeric complexes of CatK in the presence of glycosaminoglycans has been associated with its collagenase activity. Inhibitors that disrupt these complexes can selectively block the collagenase activity without interfering with the other regulatory proteolytic activities of the enzyme. Here, we have developed a fluorescence polarization (FP) assay to screen 4761 compounds for substrate-specific ectosteric collagenase inhibitors of CatK. A total of 38 compounds were identified that block the collagenase activity without interfering with the hydrolysis of active site substrates such as the synthetic peptide substrate, benzyloxycarbonyl-Phe-Arg-7-amido-4-methylcoumarin, and gelatin. The identified inhibitors can be divided into two main classes, negatively charged and polyaromatic compounds which suggest the binding to different ectosteric sites. Two of the inhibitors were highly effective in preventing the bone-resorption activity of CatK in osteoclasts. Interestingly, some of the ectosteric inhibitors were capable of differentiating between the collagenase and elastase activity of CatK depending on the ectosteric site utilized by the compound. Owing to their substrate-specific selectivity, ectosteric inhibitors represent a viable alternative to side effect-prone active site-directed inhibitors.
Collapse
|
229
|
Plotkin LI, Bruzzaniti A. Molecular signaling in bone cells: Regulation of cell differentiation and survival. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 116:237-281. [PMID: 31036293 PMCID: PMC7416488 DOI: 10.1016/bs.apcsb.2019.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The achievement of proper bone mass and architecture, and their maintenance throughout life requires the concerted actions of osteoblasts, the bone forming cells, and osteoclasts, the bone resorbing cells. The differentiation and activity of osteoblasts and osteoclasts are regulated by molecules produced by matrix-embedded osteocytes, as well as by cross talk between osteoblasts and osteoclasts through secreted factors. In addition, it is likely that direct contact between osteoblast and osteoclast precursors, and the contact of these cells with osteocytes and cells in the bone marrow, also modulates bone cell differentiation and function. With the advancement of molecular and genetic tools, our comprehension of the intracellular signals activated in bone cells has evolved significantly, from early suggestions that osteoblasts and osteoclasts have common precursors and that osteocytes are inert cells in the bone matrix, to the very sophisticated understanding of a network of receptors, ligands, intracellular kinases/phosphatases, transcription factors, and cell-specific genes that are known today. These advances have allowed the design and FDA-approval of new therapies to preserve and increase bone mass and strength in a wide variety of pathological conditions, improving bone health from early childhood to the elderly. We have summarized here the current knowledge on selected intracellular signal pathways activated in osteoblasts, osteocytes, and osteoclasts.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States.
| | - Angela Bruzzaniti
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States; Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, United States
| |
Collapse
|
230
|
Abstract
Adequate bone remodeling may be a primary parameter for long-term successful complication-free dental implant treatment. A 1.8-mm osseous thickness around dental implants is thought to be the minimum thickness for adequate vasculature for osteocyte nutrition and function. A dental implant does not provide progenitor cells or angiogenic or osteogenic factors. Thus, the surrounding bone may need to have a 1.8-mm thickness to accommodate the vasculature necessary for nutrients for appropriate remodeling. Additionally, the 1.8-mm dimension may provide for mechanical load resistance. There is no evidence to illustrate the physiologic need for the 1.8-mm dimension. This dimension requirement is based on clinical outcome observations. Basic science research for bone survival around dental implants is needed.
Collapse
|
231
|
Lee S, Prisby RD. Short-term intermittent PTH 1-34 administration and bone marrow blood vessel ossification in Mature and Middle-Aged C57BL/6 mice. Bone Rep 2019; 10:100193. [PMID: 30701186 PMCID: PMC6348201 DOI: 10.1016/j.bonr.2018.100193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 11/29/2022] Open
Abstract
Intermittent parathyroid hormone (PTH) administration augments bone and progressive bone marrow blood vessel (BMBV) ossification occurs with advancing age. Since intermittent PTH administration augments bone, it may also serve to increase BMBV ossification. We assessed the influence of 5- and 10-days of intermittent PTH 1–34 administration on trabecular and cortical bone and BMBV ossification in mature (6–8 mon; n = 30) and middle-aged (10–12 mon; n = 30) male and female C57BL/6 mice. Mice were divided accordingly: control (CON) and 5-days (5dPTH) and 10-days (10dPTH) of PTH. Mice were given PBS (50 μl) or PTH 1–34 (43 μg/kg/d) for 5- and 10-consecutive days. Trabecular bone microarchitecture (i.e., BV/TV [%], Tb.Th [μm], Tb.N [/mm], and Tb.Sp [μm]) was assessed in the distal femoral metaphysis and cortical bone parameters (i.e., Ct.Th [μm] and CSMI [mm4]) at the femoral mid-shaft. BMBV ossification (i.e., ossified vessel volume [OsVV, %] and ossified vessel thickness [OsV.Th, μm]) was assessed in the medullary cavity of the femoral shaft. All parameters were determined by μCT. At this sample size, no gender-related differences were observed so female and male data were pooled. There were no main effects nor interactions for trabecular microarchitecture and Ct.Th. However, CSMI was larger (p < 0.05) in Middle-Age vs. Mature and larger (p < 0.05) in CON and 10dPTH vs. 5dPTH. OsVV tended (p = 0.057) to be higher (0.18 ± 0.04% vs. 0.09 ± 0.02%, respectively) and OsV.Th was higher (p < 0.05; 17.4 ± 1.6 μm vs. 12.1 ± 1.4 μm, respectively) in Middle-Aged vs. Mature mice. OsVV was not altered, but ossified vessels tended (p = 0.08) to be thicker in 10dPTH (17.6 ± 2.0 μm) vs. CON (12.5 ± 1.7 μm). No interactions were observed for OsVV and OsV.Th. In conclusion, this is the first report of ossified BMBV in C57BL/6 mice. The increased OsV.Th in Middle-Aged mice coincides with previous reports of increased OsVV in aged rats. The tendency of augmented OsV.Th in 10dPTH suggests that this treatment may ultimately impair the patency of bone marrow blood vessels. Bone marrow blood vessel (BMBV) ossification occurs in rats and humans. This is the first report of BMBV ossification in Mature and Middle-Aged mice. Intermittent PTH administration tended to thicken ossified BMBV. PTH treatment may ultimately impact the patency of bone marrow blood vessels.
Collapse
Affiliation(s)
- Seungyong Lee
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX 76019, United States of America
| | - Rhonda D Prisby
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX 76019, United States of America
| |
Collapse
|
232
|
Balanta-Melo J, Toro-Ibacache V, Kupczik K, Buvinic S. Mandibular Bone Loss after Masticatory Muscles Intervention with Botulinum Toxin: An Approach from Basic Research to Clinical Findings. Toxins (Basel) 2019; 11:toxins11020084. [PMID: 30717172 PMCID: PMC6409568 DOI: 10.3390/toxins11020084] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/14/2022] Open
Abstract
The injection of botulinum toxin type A (BoNT/A) in the masticatory muscles, to cause its temporary paralysis, is a widely used intervention for clinical disorders such as oromandibular dystonia, sleep bruxism, and aesthetics (i.e., masseteric hypertrophy). Considering that muscle contraction is required for mechano-transduction to maintain bone homeostasis, it is relevant to address the bone adverse effects associated with muscle condition after this intervention. Our aim is to condense the current and relevant literature about mandibular bone loss in fully mature mammals after BoNT/A intervention in the masticatory muscles. Here, we compile evidence from animal models (mice, rats, and rabbits) to clinical studies, demonstrating that BoNT/A-induced masticatory muscle atrophy promotes mandibular bone loss. Mandibular bone-related adverse effects involve cellular and metabolic changes, microstructure degradation, and morphological alterations. While bone loss has been detected at the mandibular condyle or alveolar bone, cellular and molecular mechanisms involved in this process must still be elucidated. Further basic research could provide evidence for designing strategies to control the undesired effects on bone during the therapeutic use of BoNT/A. However, in the meantime, we consider it essential that patients treated with BoNT/A in the masticatory muscles be warned about a putative collateral mandibular bone damage.
Collapse
Affiliation(s)
- Julián Balanta-Melo
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile.
- School of Dentistry, Universidad del Valle, Cali 760043, Colombia.
- Max Planck Weizmann Center for Integrative Archaeology and Anthropology, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany.
| | - Viviana Toro-Ibacache
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile.
- Center for Quantitative Analysis in Dental Anthropology, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile.
- Department of Human Evolution, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany.
| | - Kornelius Kupczik
- Max Planck Weizmann Center for Integrative Archaeology and Anthropology, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany.
- Center for Quantitative Analysis in Dental Anthropology, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile.
| | - Sonja Buvinic
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile.
- Center for Exercise, Metabolism and Cancer Studies CEMC2016, Faculty of Medicine, Universidad de Chile, Independencia 8380453, Chile.
| |
Collapse
|
233
|
Daniele S, Giacomelli C, Pietrobono D, Barresi E, Piccarducci R, La Pietra V, Taliani S, Da Settimo F, Marinelli L, Novellino E, Martini C, Trincavelli ML. Long lasting inhibition of Mdm2-p53 interaction potentiates mesenchymal stem cell differentiation into osteoblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:737-749. [PMID: 30703414 DOI: 10.1016/j.bbamcr.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/03/2018] [Accepted: 01/24/2019] [Indexed: 12/13/2022]
Abstract
The osteoblast generation from Mesenchymal stem cells (MSCs) is tightly coordinated by transcriptional networks and signalling pathways that control gene expression and protein stability of osteogenic "master transcription factors". Among these pathways, a great attention has been focused on p53 and its physiological negative regulator, the E3 ligase Murine double minute 2 (Mdm2). Nevertheless, the signalling that regulates Mdm2-p53 axis in osteoblasts remain to be elucidated, also considering that Mdm2 possesses numerous p53-independent activities and interacts with additional proteins. Herein, the effects of Mdm2 modulation on MSC differentiation were examined by the use of short- and long-lasting inhibitors of the Mdm2-p53 complex. The long-lasting Mdm2-p53 dissociation was demonstrated to enhance the MSC differentiation into osteoblasts. The increase of Mdm2 levels promoted its association to G protein-coupled receptors kinase (GRK) 2, one of the most relevant kinases involved in the desensitization of G protein-coupled receptors (GPCRs). In turn, the long-lasting Mdm2-p53 dissociation decreased GRK2 levels and favoured the functionality of A2B Adenosine Receptors (A2BARs), a GPCR dictating MSC fate. EB148 facilitated cAMP accumulation, and mediated a sustained activation of extracellular signal-regulated kinases (ERKs) and cAMP response element-binding protein (CREB). Such pro-osteogenic effects were not detectable by using the reversible Mdm2-p53 complex inhibitor, suggesting the time course of Mdm2-p53 dissociation may impact on intracellular proteins involved in cell differentiation fate. These results suggest that the long-lasting Mdm2 binding plays a key role in the mobilization of intracellular proteins that regulate the final biological outcome of MSCs.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | | | | | | | - Valeria La Pietra
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Luciana Marinelli
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy.
| | | |
Collapse
|
234
|
Lionikaite V, Henning P, Drevinge C, Shah FA, Palmquist A, Wikström P, Windahl SH, Lerner UH. Vitamin A decreases the anabolic bone response to mechanical loading by suppressing bone formation. FASEB J 2019; 33:5237-5247. [PMID: 30668919 PMCID: PMC6436664 DOI: 10.1096/fj.201802040r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Increased vitamin A consumption is associated with decreased cortical bone mass and increased fracture risk in humans. Rodent studies have demonstrated that hypervitaminosis A increases cortical bone resorption, whereas the importance of the effects on bone formation is less well defined. We used an experimental model of increased bone formation by loading of the tibiae to investigate the effect of vitamin A on bone formation. Control [retinol activity equivalents (RAE) 4.5 µg/g chow] or vitamin A (RAE 60 µg/g chow) diets were given to female C57BL/6N mice for 4 wk, after which the tibiae were subjected to axial loading on alternate days for 2 wk, while the diets were continued. Vitamin A inhibited the loading-induced increase in trabecular and cortical bone volume. This was attributed to inhibition of loading-induced increase in osteoblast number and activity, and expression of osteoblastic genes Sp7, Alpl, and Col1a1 in cortical bone. Vitamin A, loading, and combination thereof also resulted in site-specific effects on bone composition measured by Raman spectroscopy. In summary, a clinically relevant dose of vitamin A suppresses the loading-induced gain of bone mass by decreasing bone formation. These observations may have implications for regulation of bone mass caused by physical activity and the risk of osteoporosis in humans.-Lionikaite, V., Henning, P., Drevinge, C., Shah, F. A., Palmquist, A., Wikström, P., Windahl, S. H., Lerner, U. H. Vitamin A decreases the anabolic bone response to mechanical loading by suppressing bone formation.
Collapse
Affiliation(s)
- Vikte Lionikaite
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Christina Drevinge
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Furqan A Shah
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; and
| | - Anders Palmquist
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; and
| | - Pernilla Wikström
- Department of Medical Bioscience, Pathology, Umeå University, Umeå, Sweden
| | - Sara H Windahl
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
235
|
Shin Y, Won Y, Yang JI, Chun JS. CYTL1 regulates bone homeostasis in mice by modulating osteogenesis of mesenchymal stem cells and osteoclastogenesis of bone marrow-derived macrophages. Cell Death Dis 2019; 10:47. [PMID: 30718470 PMCID: PMC6362050 DOI: 10.1038/s41419-018-1284-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 01/26/2023]
Abstract
We previously showed that mice with knockout of Cytl1, a functionally uncharacterized cytokine candidate, exhibit normal endochondral ossification and long-bone development. Here, we investigated the potential functions of CYTL1 in bone homeostasis. We found that Cytl1−/− mice exhibited higher bone mass than wild-type littermates and resisted ovariectomy-induced bone resorption. This led us to investigate the functions of CYTL1 in the osteogenesis and osteoclastogenesis of bone marrow-derived stem cells. CYTL1 was down-regulated during the osteogenesis of human mesenchymal stem cells (hMSCs). The osteogenesis of hMSCs was inhibited by overexpression or exogenous treatment of CYTL1, but enhanced by CYTL1 knockdown. CYTL1 decreased osteogenesis by inhibiting RUNX2 and promoted proliferation among undifferentiated hMSCs, but stimulated apoptosis among osteogenically differentiating cells. Finally, Cytl1−/− mice exhibited inhibition of osteoclast activity and the osteoclastogenesis of bone marrow-derived macrophages. Our results collectively suggest that CYTL1 negatively regulates the osteogenesis of MSCs and positively regulates osteoclastogenesis to modulate bone mass in mice.
Collapse
Affiliation(s)
- Youngnim Shin
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Yoonkyung Won
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Jeong-In Yang
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Jang-Soo Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea.
| |
Collapse
|
236
|
Shirakawa J, Kajikawa S, Böttcher RT, Costell M, Izu Y, Hayata T, Noda M, Ezura Y. Profilin 1 Negatively Regulates Osteoclast Migration in Postnatal Skeletal Growth, Remodeling, and Homeostasis in Mice. JBMR Plus 2019; 3:e10130. [PMID: 31346562 DOI: 10.1002/jbm4.10130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/16/2018] [Accepted: 10/21/2018] [Indexed: 01/29/2023] Open
Abstract
Profilin 1 (Pfn1), a regulator of actin polymerization, controls cell movement in a context-dependent manner. Pfn1 supports the locomotion of most adherent cells by assisting actin-filament elongation, as has been shown in skeletal progenitor cells in our previous study. However, because Pfn1 has also been known to inhibit migration of certain cells, including T cells, by suppressing branched-end elongation of actin filaments, we hypothesized that its roles in osteoclasts may be different from that of osteoblasts. By investigating the osteoclasts in culture, we first verified that Pfn1-knockdown (KD) enhances bone resorption in preosteoclastic RAW264.7 cells, despite having a comparable number and size of osteoclasts. Pfn1-KD in bone marrow cells showed similar results. Mechanistically, Pfn1-KD osteoclasts appeared more mobile than in controls. In vivo, the osteoclast-specific conditional Pfn1-deficient mice (Pfn1-cKO) by CathepsinK-Cre driver demonstrated postnatal skeletal phenotype, including dwarfism, craniofacial deformities, and long-bone metaphyseal osteolytic expansion, by 8 weeks of age. Metaphyseal and diaphyseal femurs were drastically expanded with suppressed trabecular bone mass as indicated by μCT analysis. Histologically, TRAP-positive osteoclasts were increased at endosteal metaphysis to diaphysis of Pfn1-cKO mice. The enhanced movement of Pfn1-cKO osteoclasts in culture was associated with a slight increase in cell size and podosome belt length, as well as an increase in bone-resorbing activity. Our study, for the first time, demonstrated that Pfn1 has critical roles in inhibiting osteoclast motility and bone resorption, thereby contributing to essential roles in postnatal skeletal homeostasis. Our study also provides novel insight into understanding skeletal deformities in human disorders.
Collapse
Affiliation(s)
- Jumpei Shirakawa
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Department of Oral Medicine and Stomatology School of Dental Medicine Tsurumi University Yokohama Japan
| | - Shuhei Kajikawa
- Frontier Research Unit Skeletal Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| | - Ralph T Böttcher
- Department of Molecular Medicine Max Planck Institute of Biochemistry Martinsried Germany
| | - Mercedes Costell
- Department of Biochemistry and Molecular Biology Faculty of Biology University of Valencia Spain
| | - Yayoi Izu
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| | - Tadayoshi Hayata
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Department of Molecular Pharmacology Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Science Tokyo University of Science Noda CHIBA Japan
| | - Masaki Noda
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Yokohama City Minato Red Cross Hospital Yokohama Japan.,Department of Orthopedic Surgery Tokyo Medical and Dental University Tokyo Japan
| | - Yoichi Ezura
- Department of Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan.,Frontier Research Unit Skeletal Molecular Pharmacology Medical Research Institute Tokyo Medical and Dental University Tokyo Japan
| |
Collapse
|
237
|
Yan L, Jiang J, Ma C, Li R, Xia Y. [Effect of knocking down Piezo1 mechanically sensitive protein on migration of MC3T3-E1 osteoblast cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:28-34. [PMID: 30644257 PMCID: PMC8337238 DOI: 10.7507/1002-1892.201806121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/10/2018] [Indexed: 01/22/2023]
Abstract
Objective To discuss the effect of Piezo1 mechanically sensitive protein in migration process of mouse MC3T3-E1 osteoblast cells. Methods The 5th-10th generation mouse MC3T3-E1 osteoblasts were divided into Piezo1-small interfering RNA (siRNA) transfection group (group A), negative control group (group B), and blank control group (group C). Piezo1-siRNA or negative control siRNA was transfected into mouse MC3T3-E1 osteoblasts by siRNA transfection reagent, respectively; group C was only added with siRNA transfection reagent; and the cell morphology was observed under inverted phase contrast microscope and fluorescence microscope, and the transfection efficiency was calculated. The expression of Piezo1 protein was detected by immunofluorescence staining and Western blot. Transwell cell migration assay and cell scratch assay were used to detect the migration of MC3T3-E1 osteoblasts after Piezo1-siRNA transfection. Results After 48 hours of transfection, group A showed a slight increase in cell volume and mutant growth, but cell colonies decreased, suspension cells increased and cell fragments increased when compared with untransfected cells. Under fluorescence microscope, green fluorescence was observed in MC3T3-E1 osteoblasts of group B, and the transfection efficiency was 68.56%±4.12%. Immunofluorescence staining and Western blot results showed that the expression level of Piezo1 protein in group A was significantly lower than that in groups B and C ( P<0.05); there was no significant difference between group B and group C ( P>0.05). Transwell cell migration assay and cell scratch assay showed that the number of cells per hole and the scratch healing rate of cells cultured for 1-4 days in group A were significantly lower than those in groups B and C ( P<0.05); there was no significant difference between group B and group C ( P>0.05). Conclusion Piezo1 knocked down by siRNA can inhibit the migration ability of MC3T3-E1 osteoblast cells.
Collapse
Affiliation(s)
- Liang Yan
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Jin Jiang
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Chongwen Ma
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Rui Li
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000, P.R.China
| | - Yayi Xia
- Department of Orthopedics, Gansu Key Laboratory of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, 730000,
| |
Collapse
|
238
|
Panicker LM, Srikanth MP, Castro-Gomes T, Miller D, Andrews NW, Feldman RA. Gaucher disease iPSC-derived osteoblasts have developmental and lysosomal defects that impair bone matrix deposition. Hum Mol Genet 2019; 27:811-822. [PMID: 29301038 DOI: 10.1093/hmg/ddx442] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/27/2017] [Indexed: 01/18/2023] Open
Abstract
Gaucher disease (GD) is caused by bi-allelic mutations in GBA1, the gene that encodes acid β-glucocerebrosidase (GCase). Individuals affected by GD have hematologic, visceral and bone abnormalities, and in severe cases there is also neurodegeneration. To shed light on the mechanisms by which mutant GBA1 causes bone disease, we examined the ability of human induced pluripotent stem cells (iPSC) derived from patients with Types 1, 2 and 3 GD, to differentiate to osteoblasts and carry out bone deposition. Differentiation of GD iPSC to osteoblasts revealed that these cells had developmental defects and lysosomal abnormalities that interfered with bone matrix deposition. Compared with controls, GD iPSC-derived osteoblasts exhibited reduced expression of osteoblast differentiation markers, and bone matrix protein and mineral deposition were defective. Concomitantly, canonical Wnt/β catenin signaling in the mutant osteoblasts was downregulated, whereas pharmacological Wnt activation with the GSK3β inhibitor CHIR99021 rescued GD osteoblast differentiation and bone matrix deposition. Importantly, incubation with recombinant GCase (rGCase) rescued the differentiation and bone-forming ability of GD osteoblasts, demonstrating that the abnormal GD phenotype was caused by GCase deficiency. GD osteoblasts were also defective in their ability to carry out Ca2+-dependent exocytosis, a lysosomal function that is necessary for bone matrix deposition. We conclude that normal GCase enzymatic activity is required for the differentiation and bone-forming activity of osteoblasts. Furthermore, the rescue of bone matrix deposition by pharmacological activation of Wnt/β catenin in GD osteoblasts uncovers a new therapeutic target for the treatment of bone abnormalities in GD.
Collapse
Affiliation(s)
- Leelamma M Panicker
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Manasa P Srikanth
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thiago Castro-Gomes
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD 20742, USA
| | - Diana Miller
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Norma W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD 20742, USA
| | - Ricardo A Feldman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
239
|
Essex AL, Pin F, Huot JR, Bonewald LF, Plotkin LI, Bonetto A. Bisphosphonate Treatment Ameliorates Chemotherapy-Induced Bone and Muscle Abnormalities in Young Mice. Front Endocrinol (Lausanne) 2019; 10:809. [PMID: 31803146 PMCID: PMC6877551 DOI: 10.3389/fendo.2019.00809] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy is frequently accompanied by several side effects, including nausea, diarrhea, anorexia and fatigue. Evidence from ours and other groups suggests that chemotherapy can also play a major role in causing not only cachexia, but also bone loss. This complicates prognosis and survival among cancer patients, affects quality of life, and can increase morbidity and mortality rates. Recent findings suggest that soluble factors released from resorbing bone directly contribute to loss of muscle mass and function secondary to metastatic cancer. However, it remains unknown whether similar mechanisms also take place following treatments with anticancer drugs. In this study, we found that young male CD2F1 mice (8-week old) treated with the chemotherapeutic agent cisplatin (2.5 mg/kg) presented marked loss of muscle and bone mass. Myotubes exposed to bone conditioned medium from cisplatin-treated mice showed severe atrophy (-33%) suggesting a bone to muscle crosstalk. To test this hypothesis, mice were administered cisplatin in combination with an antiresorptive drug to determine if preservation of bone mass has an effect on muscle mass and strength following chemotherapy treatment. Mice received cisplatin alone or combined with zoledronic acid (ZA; 5 μg/kg), a bisphosphonate routinely used for the treatment of osteoporosis. We found that cisplatin resulted in progressive loss of body weight (-25%), in line with reduced fat (-58%) and lean (-17%) mass. As expected, microCT bone histomorphometry analysis revealed significant reduction in bone mass following administration of chemotherapy, in line with reduced trabecular bone volume (BV/TV) and number (Tb.N), as well as increased trabecular separation (Tb.Sp) in the distal femur. Conversely, trabecular bone was protected when cisplatin was administered in combination with ZA. Interestingly, while the animals exposed to chemotherapy presented significant muscle wasting (~-20% vs. vehicle-treated mice), the administration of ZA in combination with cisplatin resulted in preservation of muscle mass (+12%) and strength (+42%). Altogether, these observations support our hypothesis of bone factors targeting muscle and suggest that pharmacological preservation of bone mass can benefit muscle mass and function following chemotherapy.
Collapse
Affiliation(s)
- Alyson L. Essex
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Joshua R. Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lynda F. Bonewald
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- IUPUI Center for Cachexia Research, Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lilian I. Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrea Bonetto
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
- IUPUI Center for Cachexia Research, Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Otolaryngology – Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Andrea Bonetto
| |
Collapse
|
240
|
Koide M, Kobayashi Y. Regulatory mechanisms of sclerostin expression during bone remodeling. J Bone Miner Metab 2019; 37:9-17. [PMID: 30357564 DOI: 10.1007/s00774-018-0971-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/14/2018] [Indexed: 11/28/2022]
Abstract
Osteocytes are embedded in bone matrices and are connected to each other to respond to mechanical loading on bone. Recent studies have demonstrated the roles of mechanical loading in bone accrual. Bone responds to mechanical loading by decreasing the expression of sclerostin, an inhibitor of Wnt/β-catenin signals, in osteocytes. This increases bone mass because the activation of Wnt/β-catenin signals in bone microenvironments promotes bone formation and suppresses bone resorption. Thus, in recent years, sclerostin have attracted increasing attention in bone metabolism. However, the regulatory mechanism of sclerostin expression during bone remodeling has not been fully elucidated. In this review, we summarized the regulation of bone formation and resorption by Wnt signals, a Wnt/β-catenin signal inhibitor sclerostin, and molecular mechanisms by which the expression of sclerostin is suppressed by mechanical loading and parathyroid hormone. We also discuss a possibility that osteoclasts suppress the expression of sclerostin during bone remodeling, which in turn, promote bone formation. The effectiveness of an anti-sclerostin antibody with anti-dickkopf-1 antibody for increasing bone mass was discussed.
Collapse
Affiliation(s)
- Masanori Koide
- Division of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri, Nagano, 399-0781, Japan
| | - Yasuhiro Kobayashi
- Division of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri, Nagano, 399-0781, Japan.
| |
Collapse
|
241
|
Qiao J, Liu A, Liu J, Guan D, Chen T. Salvianolic acid B (Sal B) alleviates the decreased activity induced by prednisolone acetate on osteoblasts by up-regulation of bone formation and differentiation genes. Food Funct 2019; 10:6184-6192. [PMID: 31501830 DOI: 10.1039/c9fo01246j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sal B could promote bone formation and help protect against bone loss caused by prednisolone acetate treatment.
Collapse
Affiliation(s)
- Jiutao Qiao
- The Second Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Aiyun Liu
- The Second Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Jianyu Liu
- The Second Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Dehong Guan
- The Second Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Tianxin Chen
- The Second Affiliated Hospital of Harbin Medical University
- Harbin
- China
| |
Collapse
|
242
|
Baumhoer D, Amary F, Flanagan AM. An update of molecular pathology of bone tumors. Lessons learned from investigating samples by next generation sequencing. Genes Chromosomes Cancer 2018; 58:88-99. [PMID: 30582658 DOI: 10.1002/gcc.22699] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/25/2018] [Accepted: 10/25/2018] [Indexed: 12/27/2022] Open
Abstract
The last decade has seen the majority of primary bone tumor subtypes become defined by molecular genetic alteration. Examples include giant cell tumour of bone (H3F3A p.G34W), chondroblastoma (H3F3B p.K36M), mesenchymal chondrosarcoma (HEY1-NCOA2), chondromyxoid fibroma (GRM1 rearrangements), aneurysmal bone cyst (USP6 rearrangements), osteoblastoma/osteoid osteoma (FOS/FOSB rearrangements), and synovial chondromatosis (FN1-ACVR2A and ACVR2A-FN1). All such alterations are mutually exclusive. Many of these have been translated into clinical service using immunohistochemistry or FISH. 60% of central chondrosarcoma is characterised by either isocitrate dehydrogenase (IDH) 1 or IDH2 mutations distinguishing them from other cartilaginous tumours. In contrast, recurrent alterations which are clinically helpful have not been found in high grade osteosarcoma. High throughput next generation sequencing has also proved valuable in identifying germ line alterations in a significant proportion of young patients with primary malignant bone tumors. These findings will play an increasing role in reaching a diagnosis and in patient management.
Collapse
Affiliation(s)
- Daniel Baumhoer
- Bone Tumour Reference Centre, Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Fernanda Amary
- Department of Pathology, The Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom.,Department of Pathology, Cancer Institute, University College London, London, United Kingdom
| | - Adrienne M Flanagan
- Department of Pathology, The Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom.,Department of Pathology, Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
243
|
Suzuki T, Nakamura Y, Kato H. Calcium and vitamin D supplementation with 3-year denosumab treatment is beneficial to enhance bone mineral density in postmenopausal patients with osteoporosis and rheumatoid arthritis. Ther Clin Risk Manag 2018; 15:15-22. [PMID: 30588001 PMCID: PMC6302805 DOI: 10.2147/tcrm.s182858] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background This 3-year retrospective study compared the outcomes of bisphosphonate-pretreated denosumab therapy with or without vitamin D and calcium supplementation in postmenopausal osteoporosis (OP) patients with rheumatoid arthritis (RA). Materials and methods Fifty-eight patients under long-term denosumab treatment were divided into groups without (denosumab group; 31 cases) or with (combination group; 27 cases) vitamin D and calcium supplementation. The bone markers of BAP, TRACP-5b, and urinary NTX were measured at baseline and every year for 3 years. We also evaluated bone mineral density (BMD) of the lumbar 1–4 vertebrae (L-BMD) and bilateral total hips (H-BMD) at the same time points. Results There were no significant differences in the percent changes of serum albumin-corrected calcium between the groups. The percent change in TRACP-5b was significantly higher in the combination group at 2 years. Serum 25-hydroxyvitamin D status was persistently high during therapy in both groups, with significant percent increases over baseline at 2 and 6 months in both groups and at 24 months in the combination group. The percent increase from baseline of serum zinc was significantly higher at 3 years in the combination group over the denosumab group. L-BMD and H-BMD were significantly increased at every time point for 3 years vs pretreatment levels in both groups and were significantly higher in the combination group at all time points. Conclusion Compared with denosumab monotherapy, the combination group displayed significantly increased serum zinc, L-BMD, and H-BMD at 3 years in OP patients with RA. Thus, calcium and vitamin D supplementation may be beneficial to enhance BMD gains, but not necessarily 25-hydroxyvitamin D status, in patients with OP and RA under denosumab.
Collapse
Affiliation(s)
- Takako Suzuki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan,
| | - Yukio Nakamura
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan, .,Department of Orthopaedic Surgery, Showa Inan General Hospital, Komagane, Japan,
| | - Hiroyuki Kato
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan,
| |
Collapse
|
244
|
Atkinson SP. A Preview of Selected Articles. Stem Cells Transl Med 2018. [DOI: 10.1002/sctm.18-0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
245
|
Huang S, Li Z, Liu Y, Gao D, Zhang X, Hao J, Yang F. Neural regulation of bone remodeling: Identifying novel neural molecules and pathways between brain and bone. J Cell Physiol 2018; 234:5466-5477. [PMID: 29377116 DOI: 10.1002/jcp.26502] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 01/20/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Shishu Huang
- Department of Orthopaedic Surgery West China Hospital, Sichuan University Chengdu China
| | - Zhenxia Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics West China Hospital of Stomatology, Sichuan University Chengdu China
| | - Yunhui Liu
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Dashuang Gao
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Xinzhou Zhang
- Department of Nephrology Shenzhen People's Hospital, Second Clinical Medical College, Jinan University Shenzhen China
| | - Jin Hao
- Program in Biological Sciences in Dental Medicine, Harvard School of Dental Medicine Boston Massachusetts
| | - Fan Yang
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| |
Collapse
|
246
|
Fornetti J, Welm AL, Stewart SA. Understanding the Bone in Cancer Metastasis. J Bone Miner Res 2018; 33:2099-2113. [PMID: 30476357 DOI: 10.1002/jbmr.3618] [Citation(s) in RCA: 280] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 12/11/2022]
Abstract
The bone is the third most common site of metastasis for a wide range of solid tumors including lung, breast, prostate, colorectal, thyroid, gynecologic, and melanoma, with 70% of metastatic prostate and breast cancer patients harboring bone metastasis.1 Unfortunately, once cancer spreads to the bone, it is rarely cured and is associated with a wide range of morbidities including pain, increased risk of fracture, and hypercalcemia. This fact has driven experts in the fields of bone and cancer biology to study the bone, and has revealed that there is a great deal that each can teach the other. The complexity of the bone was first described in 1889 when Stephen Paget proposed that tumor cells have a proclivity for certain organs, where they "seed" into a friendly "soil" and eventually grow into metastatic lesions. Dr. Paget went on to argue that although many study the "seed" it would be paramount to understand the "soil." Since this original work, significant advances have been made not only in understanding the cell-autonomous mechanisms that drive metastasis, but also alterations which drive changes to the "soil" that allow a tumor cell to thrive. Indeed, it is now clear that the "soil" in different metastatic sites is unique, and thus the mechanisms that allow tumor cells to remain in a dormant or growing state are specific to the organ in question. In the bone, our knowledge of the components that contribute to this fertile "soil" continues to expand, but our understanding of how they impact tumor growth in the bone remains in its infancy. Indeed, we now appreciate that the endosteal niche likely contributes to tumor cell dormancy, and that osteoclasts, osteocytes, and adipocytes can impact tumor cell growth. Here, we discuss the bone microenvironment and how it impacts cancer cell seeding, dormancy, and growth. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jaime Fornetti
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Sheila A Stewart
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.,Integrating Communication within the Cancer Environment (ICCE) Institute, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
247
|
Lee JE, Lee H, Kim MH, Yang WM. Osteogenic effects of Phlomis umbrosa via up-regulation of Runx2 in osteoporosis. Biomed Rep 2018; 10:17-22. [PMID: 30588298 PMCID: PMC6299205 DOI: 10.3892/br.2018.1172] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/07/2018] [Indexed: 01/21/2023] Open
Abstract
Phlomis umbrosa Turcz (labiatae) has been suggested to promote bone growth. However, the anti-osteoporotic effects of P. umbrosa have not yet been elucidated. In the present study, the osteogenic effects of P. umbrosa were investigated in an osteoporosis model. ICR female mice were ovariectomized (OVX) to induce osteoporosis for 7 weeks. Treatment with 1, 10 and 100 mg/kg P. umbrosa was administrated orally to the OVX mice for 6 weeks. At the end of experiment, the microstructure of the capital femoral epiphysis was investigated. The levels of bone mineral density (BMD), bone mineral content (BMC) and serum osteocalcin concentration were evaluated. In addition, mineralized Saos-2 osteoblast cells were treated with 0.01, 0.1 and 1 µg/ml P. umbrosa to analyze the expression of osteoblast differentiation-associated factors. Hyperplasia of the growth plate in the femur was recovered by P. umbrosa treatment. BMD and BMC were significantly increased in P. umbrosa-treated femurs. Serum calcium concentration was increased following P. umbrosa treatment. In addition, the ratio of mineralization was markedly increased in P. umbrosa-treated differentiated osteoblasts along with increases in Runx2 levels. P. umbrosa conferred its osteogenic effects by upregulating Runx2 in osteoporosis. P. umbrosa may be a potential therapeutic material for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Ji Eun Lee
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Haesu Lee
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mi Hye Kim
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woong Mo Yang
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
248
|
NUMB maintains bone mass by promoting degradation of PTEN and GLI1 via ubiquitination in osteoblasts. Bone Res 2018; 6:32. [PMID: 30455992 PMCID: PMC6226489 DOI: 10.1038/s41413-018-0030-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/22/2018] [Accepted: 03/13/2018] [Indexed: 02/05/2023] Open
Abstract
The adaptor protein NUMB is involved in asymmetric division and cell fate determination and recognized as an antagonist of Notch. Previous studies have proved that Notch activation in osteoblasts contributes to a high bone mass. In this study, however, an osteopenic phenotype was found in 9-week-old mice using osteoblastic specific Col1a1–2.3-Cre to ablate both Numb and its homologue Numbl . The trabecular bone mass decreased dramatically while the cortical bone mass was unaffected. Here, the Notch signal was not activated, while the tensin homologue deleted on human chromosome 10 (PTEN), which dephosphorylates phosphatidylinositide 3-kinases, was elevated, attenuating protein kinase B (Akt). The ubiquitination assay revealed that NUMB may physiologically promote PTEN ubiquitination in the presence of neural precursor cell-expressed developmentally downregulated protein 4–1. In addition, the deficiency of Numb/Numbl also activated the Hedgehog pathway through GLI1. This process was found to improve the ratio of the receptor activator of nuclear factor-kB ligand to osteoprotegerin, which enhanced the differentiation of osteoclasts and bone resorption . In conclusion, this study provides an insight into new functons of NUMB and NUMBL on bone homeostasis. The related proteins NUMB and NUMBL maintain the survival of bone-generating osteoblast cells. NUMB was previously recognized to antagonize Notch signaling pathway ; In this study, it observes that genetically altered mice, unable to express the two proteins, suffered from degraded bone quality. This suggests that the two proteins play a more complex, nuanced role in the process of bone mass maintenance. The team’s studies showed that NUMB and NUMBL suppression inhibits a signaling pathway important to skeletal development and protein synthesis in osteoblasts, though raise that further investigations are essential to elucidate the exact mechanistic action of these proteins. The authors of this study suggest that NUMB constitutes a potential target for therapies targeting bone loss and reduced bone strength in patients with osteoporosis.
Collapse
|
249
|
Hang K, Ye C, Chen E, Zhang W, Xue D, Pan Z. Role of the heat shock protein family in bone metabolism. Cell Stress Chaperones 2018; 23:1153-1164. [PMID: 30187197 PMCID: PMC6237693 DOI: 10.1007/s12192-018-0932-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/11/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022] Open
Abstract
Heat shock proteins (HSPs) are a family of proteins produced by cells in response to exposure to stressful conditions. In addition to their role as chaperones, they also play an important role in the cardiovascular, immune, and other systems. Normal bone tissue is maintained by bone metabolism, particularly by the balance between osteoblasts and osteoclasts, which are physiologically regulated by multiple hormones and cytokines. In recent years, studies have reported the vital role of HSPs in bone metabolism. However, the conclusions remain largely controversial, and the exact mechanisms are still unclear, so a review and analyses of previous studies are of importance. This article reviews the current understanding of the roles and effects of HSPs on bone cells (osteoblasts, osteoclasts, and osteocytes), in relation to bone metabolism.
Collapse
Affiliation(s)
- Kai Hang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
- Orthopedics Research Institute, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
| | - Chenyi Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
- Orthopedics Research Institute, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
| | - Erman Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
- Orthopedics Research Institute, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
| | - Wei Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
- Orthopedics Research Institute, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
| | - Deting Xue
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
- Orthopedics Research Institute, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
| | - Zhijun Pan
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
- Orthopedics Research Institute, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009 China
| |
Collapse
|
250
|
Kuroyanagi G, Adapala NS, Yamaguchi R, Kamiya N, Deng Z, Aruwajoye O, Kutschke M, Chen E, Jo C, Ren Y, Kim HKW. Interleukin-6 deletion stimulates revascularization and new bone formation following ischemic osteonecrosis in a murine model. Bone 2018; 116:221-231. [PMID: 30125727 DOI: 10.1016/j.bone.2018.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 08/11/2018] [Accepted: 08/14/2018] [Indexed: 12/28/2022]
Abstract
Legg-Calvé-Perthes disease (LCPD) is a childhood form of ischemic osteonecrosis of the femoral head which can produce a permanent femoral head deformity and early osteoarthritis. The femoral head deformity results from increased bone resorption and decreased bone formation during repair and remodeling of the necrotic femoral head. A recent study showed that a pro-inflammatory cytokine, interleukin-6 (IL-6), is significantly elevated in the synovial fluid of patients with LCPD. We hypothesized that IL-6 elevation decreases bone formation during the repair process following ischemic osteonecrosis and that IL-6 depletion will increase new bone formation. To test this hypothesis, we surgically induced ischemic osteonecrosis in the wild-type (n = 29) and IL-6 knockout (KO) mice (n = 25). The animals were assessed at 48 h, 2 weeks and 4 weeks following the induction of ischemic osteonecrosis using histologic, histomorphometric and micro-CT methods. IL-6 immunohistochemistry showed high expression of IL-6 in the osteonecrotic side of the wild-type mice at 48 h and 4 weeks following ischemic osteonecrosis, but not in the IL-6 KO mice. We also confirmed an undetectable level of IL-6 expression in the primary osteoblasts of the IL-6 KO mice compared to the readily detectable level in the wild-type mice. Furthermore, we confirmed that IL-6 deletion did not affect the extent of bone necrosis in the IL-6 KO mice compared to the wild-type mice by performing histologic and terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling (TUNEL) assessments at 2 weeks following the induction of ischemia. Both groups had the same extent of ischemic osteonecrosis and absence of repair at 2 weeks. At 4 weeks, the necrotic epiphyses showed a significant increase in the extent of revascularization in the IL-6 KO mice compared to the wild-type mice (p = 0.001). In addition, a significantly greater recovery of the hematopoietic bone marrow was observed in the osteonecrotic side of the IL-6 KO mice compared to the wild-type mice (p < 0.01). Vascular endothelial growth factor (VEGF) immunohistochemistry showed regionally increased staining in the areas of repair in the osteonecrosis side of IL-6 KO mice compared to the wild-type mice at 4 weeks following ischemic osteonecrosis. Micro-CT assessment of the wild-type mice at 4 weeks showed a significant decrease in the percent bone volume (p < 0.01) in the osteonecrotic side compared to the control side. In contrast, IL-6 KO mice showed significantly increased bone volume in the osteonecrotic side compared to the osteonecrotic side of WT mice (p < 0.001). No significant difference in the bone volume percentage was found between the control side of the wild-type and the IL-6 KO mice. Histomorphometric analysis at 4 weeks revealed increased osteoblast number/bone surface (p < 0.001), bone formation rate (BFR) (p = 0.0001), and mineral apposition rate (MAR) (p < 0.0001) in the osteonecrotic side of the IL-6 KO mice compared to the wild-type mice. The number of osteoclast/bone surface was also increased in the IL-6 KO mice compared to the wild-type mice (p < 0.0001). No significant difference was observed between the control side of the wild-type and IL-6 KO mice with regards to the number of osteoblast or osteoclast/bone surface, BFR, and MAR. We next obtained primary osteoblasts from IL-6 KO mice and showed they expressed a significantly higher level of RANKL/OPG than wild-type mice (p = 0.001) in hypoxia culture condition. Taken together, the findings indicate that IL-6 deletion stimulates revascularization and new bone formation following ischemic osteonecrosis. This study provides new evidence that therapeutic strategies to block IL-6 may be beneficial for bone healing following ischemic osteonecrosis.
Collapse
Affiliation(s)
- Gen Kuroyanagi
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Naga Suresh Adapala
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | - Ryosuke Yamaguchi
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Department of Orthopaedic and Spine Surgery, Fukuoka Children's Hospital, Fukuoka 813-0017, Japan
| | - Nobuhiro Kamiya
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Sports Medicine, Tenri University, Tenri 632-0071, Japan
| | - Zhuo Deng
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | - Olumide Aruwajoye
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | - Michael Kutschke
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | - Elena Chen
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | - Chanhee Jo
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | - Yinshi Ren
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA
| | - Harry K W Kim
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390-8883, USA.
| |
Collapse
|