101
|
Molagoda IMN, Kang CH, Lee MH, Choi YH, Lee CM, Lee S, Kim GY. Fisetin promotes osteoblast differentiation and osteogenesis through GSK-3β phosphorylation at Ser9 and consequent β-catenin activation, inhibiting osteoporosis. Biochem Pharmacol 2021; 192:114676. [PMID: 34256044 DOI: 10.1016/j.bcp.2021.114676] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023]
Abstract
Fisetin is a bioactive flavonol that inhibits osteoclastogenesis and promotes osteoblastogenesis. However, the osteogenic activity of fisetin needs to be comprehensively elucidated. In the present study, we observed that fisetin significantly increased alkaline phosphatase (ALP) activity and bone mineralization in MC3T3-E1 preosteoblasts, accompanied by a significant increase in runt-related transcription factor 2 (RUNX2), ALP, collagen type Ⅰ alpha 1 (Col1α1), osterix (OSX), osteocalcin (OCN), and bone morphogenetic protein 4 (BMP4) expression. Furthermore, fisetin promoted vertebral formation in zebrafish larvae, with the highest fisetin concentration comparable with that observed in β-glycerophosphate treatment. Fisetin also inhibited prednisolone (PDS)-induced anti-osteoblastic genes, including nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), tartrate-resistant acid phosphatase-6 (ACP6), dendritic cell-specific transmembrane protein (DC-STAMP), and cathepsin K (CTSK). Fisetin potently mitigated the PDS-induced inhibition of ALP activity and bone mineralization, as well as vertebral resorption in zebrafish larvae. Moreover, we confirmed that fisetin-induced osteogenic effect was activated through phosphorylation of glycogen synthase kinase-3β (GSK-3β) at Ser9, consequently releasing β-catenin from the destructive complex to promote its nuclear translocation. β-Catenin inhibition by FH535 and the stabilization of GSK-3β by DOI hydrochloride remarkably inhibited fisetin-induced osteogenic activities, indicating that the GSK-3β/β-catenin signaling pathway plays a vital role in fisetin-induced osteogenesis. Collectively, our findings suggest that fisetin stimulates osteogenic activity and could be used as an effective strategy to prevent bone resorption.
Collapse
Affiliation(s)
- Ilandarage Menu Neelaka Molagoda
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Research Institute for Basic Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Chang-Hee Kang
- Nakdongggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Mi-Hwa Lee
- Nakdongggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, RI 02912, USA
| | - Seungheon Lee
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea; Research Institute for Basic Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
102
|
Wang K, Ren Y, Lin S, Jing Y, Ma C, Wang J, Yuan XB, Han X, Zhao H, Wang Z, Zheng M, Xiao Y, Chen L, Olsen BR, Feng JQ. Osteocytes but not osteoblasts directly build mineralized bone structures. Int J Biol Sci 2021; 17:2430-2448. [PMID: 34326685 PMCID: PMC8315029 DOI: 10.7150/ijbs.61012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/21/2021] [Indexed: 02/05/2023] Open
Abstract
Bone-forming osteoblasts have been a cornerstone of bone biology for more than a century. Most research toward bone biology and bone diseases center on osteoblasts. Overlooked are the 90% of bone cells, called osteocytes. This study aims to test the hypothesis that osteocytes but not osteoblasts directly build mineralized bone structures, and that defects in osteocytes lead to the onset of hypophosphatemia rickets. The hypothesis was tested by developing and modifying multiple imaging techniques, including both in vivo and in vitro models plus two types of hypophosphatemia rickets models (Dmp1-null and Hyp, Phex mutation mice), and Dmp1-Cre induced high level of β-catenin models. Our key findings were that osteocytes (not osteoblasts) build bone similar to the construction of a high-rise building, with a wire mesh frame (i.e., osteocyte dendrites) and cement (mineral matrices secreted from osteocytes), which is a lengthy and slow process whose mineralization direction is from the inside toward the outside. When osteoblasts fail to differentiate into osteocytes but remain highly active in Dmp-1-null or Hyp mice, aberrant and poor bone mineralization occurs, caused by a sharp increase in Wnt-β-catenin signaling. Further, the constitutive expression of β-catenin in osteocytes recaptures a similar osteomalacia phenotype as shown in Dmp1 null or Hyp mice. Thus, we conclude that osteocytes directly build bone, and osteoblasts with a short life span serve as a precursor to osteocytes, which challenges the existing dogma.
Collapse
Affiliation(s)
- Ke Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Yinshi Ren
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219 USA
| | - Shuxian Lin
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Shanghai, 200092, China
| | - Yan Jing
- Department of Orthodontics, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Chi Ma
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219 USA
| | - Jun Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - X Baozhi Yuan
- Angitia Biopharmaceuticals, Guangzhou, 510000, China
| | - Xianglong Han
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hu Zhao
- Department of Restorative Dentistry, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Zheng Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Minghao Zheng
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, 6009, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia
| | - Lin Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Bjorn Reino Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| |
Collapse
|
103
|
Melnik BC, Stremmel W, Weiskirchen R, John SM, Schmitz G. Exosome-Derived MicroRNAs of Human Milk and Their Effects on Infant Health and Development. Biomolecules 2021; 11:biom11060851. [PMID: 34200323 PMCID: PMC8228670 DOI: 10.3390/biom11060851] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple biologically active components of human milk support infant growth, health and development. Milk provides a wide spectrum of mammary epithelial cell-derived extracellular vesicles (MEVs) for the infant. Although the whole spectrum of MEVs appears to be of functional importance for the growing infant, the majority of recent studies report on the MEV subfraction of milk exosomes (MEX) and their miRNA cargo, which are in the focus of this review. MEX and the dominant miRNA-148a play a key role in intestinal maturation, barrier function and suppression of nuclear factor-κB (NF-κB) signaling and may thus be helpful for the prevention and treatment of necrotizing enterocolitis. MEX and their miRNAs reach the systemic circulation and may impact epigenetic programming of various organs including the liver, thymus, brain, pancreatic islets, beige, brown and white adipose tissue as well as bones. Translational evidence indicates that MEX and their miRNAs control the expression of global cellular regulators such as DNA methyltransferase 1-which is important for the up-regulation of developmental genes including insulin, insulin-like growth factor-1, α-synuclein and forkhead box P3-and receptor-interacting protein 140, which is important for the regulation of multiple nuclear receptors. MEX-derived miRNA-148a and miRNA-30b may stimulate the expression of uncoupling protein 1, the key inducer of thermogenesis converting white into beige/brown adipose tissue. MEX have to be considered as signalosomes derived from the maternal lactation genome emitted to promote growth, maturation, immunological and metabolic programming of the offspring. Deeper insights into milk's molecular biology allow the conclusion that infants are both "breast-fed" and "breast-programmed". In this regard, MEX miRNA-deficient artificial formula is not an adequate substitute for breastfeeding, the birthright of all mammals.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany;
- Correspondence: ; Tel.: +49-5241-988060
| | - Wolfgang Stremmel
- Private Praxis for Internal Medicine, Beethovenstraße 2, D-76530 Baden-Baden, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany;
- Institute for Interdisciplinary Dermatological Prevention and Rehabilitation (iDerm), University of Osnabrück, D-49076 Osnabrück, Germany
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
104
|
Chitosan-based 3D-printed scaffolds for bone tissue engineering. Int J Biol Macromol 2021; 183:1925-1938. [PMID: 34097956 DOI: 10.1016/j.ijbiomac.2021.05.215] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Despite the spontaneous regenerative properties of autologous bone grafts, this technique remains dilatory and restricted to fractures and injuries. Conventional grafting strategies used to treat bone tissue damage have several limitations. This highlights the need for novel approaches to overcome the persisting challenges. Tissue-like constructs that can mimic natural bone structurally and functionally represent a promising strategy. Bone tissue engineering (BTE) is an approach used to develop bioengineered bone with subtle architecture. BTE utilizes biomaterials to accommodate cells and deliver signaling molecules required for bone rejuvenation. Among the various techniques available for scaffold creation, 3D-printing technology is considered to be a superior technique as it enables the design of functional scaffolds with well-defined customizable properties. Among the biomaterials obtained from natural, synthetic, or ceramic origins, naturally derived chitosan (CS) polymers are promising candidates for fabricating reliable tissue constructs. In this review, the physicochemical-biological properties and applications of CS-based 3D-printed scaffolds and their future perspectives in BTE are summarized.
Collapse
|
105
|
Venetis K, Piciotti R, Sajjadi E, Invernizzi M, Morganti S, Criscitiello C, Fusco N. Breast Cancer with Bone Metastasis: Molecular Insights and Clinical Management. Cells 2021; 10:cells10061377. [PMID: 34199522 PMCID: PMC8229615 DOI: 10.3390/cells10061377] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the remarkable advances in the diagnosis and treatment of breast cancer patients, the presence or development of metastasis remains an incurable condition. Bone is one of the most frequent sites of distant dissemination and negatively impacts on patient's survival and overall frailty. The interplay between tumor cells and the bone microenvironment induces bone destruction and tumor progression. To date, the clinical management of bone metastatic breast cancer encompasses anti-tumor systemic therapies along with bone-targeting agents, aimed at slowing bone resorption to reduce the risk of skeletal-related events. However, their effect on patients' survival remains controversial. Unraveling the biology that governs the interplay between breast neoplastic cells and bone tissue would provide means for the development of new therapeutic agents. This article outlines the state-of-the art in the characterization and targeting the bone metastasis in breast cancer, focusing on the major clinical and translational studies on this clinically relevant topic.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (K.V.); (R.P.); (E.S.); (S.M.)
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Roberto Piciotti
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (K.V.); (R.P.); (E.S.); (S.M.)
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elham Sajjadi
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (K.V.); (R.P.); (E.S.); (S.M.)
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Marco Invernizzi
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy;
- Infrastruttura Ricerca Formazione Innovazione (IRFI), Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Stefania Morganti
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (K.V.); (R.P.); (E.S.); (S.M.)
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (K.V.); (R.P.); (E.S.); (S.M.)
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Correspondence: (C.C.); (N.F.); Tel.: +39-02-9437-2079 (N.F.)
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy; (K.V.); (R.P.); (E.S.); (S.M.)
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Correspondence: (C.C.); (N.F.); Tel.: +39-02-9437-2079 (N.F.)
| |
Collapse
|
106
|
Chen F, Han Y, Kang Y. Bone marrow niches in the regulation of bone metastasis. Br J Cancer 2021; 124:1912-1920. [PMID: 33758331 PMCID: PMC8184962 DOI: 10.1038/s41416-021-01329-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 02/06/2021] [Accepted: 02/16/2021] [Indexed: 12/16/2022] Open
Abstract
The bone marrow has been widely recognised to host a unique microenvironment that facilitates tumour colonisation. Bone metastasis frequently occurs in the late stages of malignant diseases such as breast, prostate and lung cancers. The biology of bone metastasis is determined by tumour-cell-intrinsic traits as well as their interaction with the microenvironment. The bone marrow is a dynamic organ in which various stages of haematopoiesis, osteogenesis, osteolysis and different kinds of immune response are precisely regulated. These different cellular components constitute specialised tissue microenvironments-niches-that play critical roles in controlling tumour cell colonisation, including initial seeding, dormancy and outgrowth. In this review, we will dissect the dynamic nature of the interactions between tumour cells and bone niches. By targeting certain steps of tumour progression and crosstalk with the bone niches, the development of potential therapeutic approaches for the clinical treatment of bone metastasis might be feasible.
Collapse
Affiliation(s)
- Fenfang Chen
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yujiao Han
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA.
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
107
|
Bushinsky DA, Raggi P, Bover J, Ketteler M, Bellasi A, Rodriguez M, Sinha S, Garg R, Perelló J, Gold A, Chertow GM. Effects of Myo-inositol Hexaphosphate (SNF472) on Bone Mineral Density in Patients Receiving Hemodialysis: An Analysis of the Randomized, Placebo-Controlled CaLIPSO Study. Clin J Am Soc Nephrol 2021; 16:736-745. [PMID: 33835939 PMCID: PMC8259477 DOI: 10.2215/cjn.16931020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/22/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES In the CaLIPSO study, intravenous administration of SNF472 (300 or 600 mg) during hemodialysis significantly attenuated progression of coronary artery and aortic valve calcification. SNF472 selectively inhibits formation of hydroxyapatite, the final step in cardiovascular calcification. Because bone mineral is predominantly hydroxyapatite, we assessed changes in bone mineral density in CaLIPSO. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Patients with coronary artery calcification at screening (Agatston score of 100-3500 U) were randomized 1:1:1 to receive placebo, 300 mg SNF472, or 600 mg SNF472 as an intravenous infusion during hemodialysis three times weekly for 52 weeks. Dual-energy x-ray absorptiometry (DXA) scans were obtained at baseline (screening) and end of treatment, and between-group changes from baseline were compared using analysis of covariance. RESULTS Among 274 randomized patients, 202 had evaluable DXA scans at baseline and postrandomization (the DXA-modified intention-to-treat population). Mean (95% confidence interval) changes in total-hip bone mineral density from baseline to week 52 were -1.5% (-2.7% to -0.3%), -1.5% (-2.7% to -0.4%), and -2.5% (-3.8% to -1.2%) in the placebo, 300 mg SNF472, and 600 mg SNF472 groups, respectively. Mean (95% confidence interval) changes in femoral-neck bone mineral density from baseline to week 52 were -0.3% (-1.6% to 1.0%), -1.0% (-2.3% to 0.2%), and -2.6% (-4.0% to -1.3%), respectively. Regression analyses showed no correlation between change in coronary artery calcium volume and change in bone mineral density at either location. Changes in serum alkaline phosphatase, calcium, magnesium, phosphate, and intact parathyroid hormone levels were similar across treatment groups. Clinical fracture events were reported for four of 90, three of 92, and six of 91 patients in the placebo, 300 mg SNF472, and 600 mg SNF472 groups, respectively. CONCLUSIONS Bone mineral density decreased modestly in all groups over 1 year. In the 600 mg SNF472 group, the reduction appeared more pronounced. Reported fractures were infrequent in all groups. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Effect of SNF472 on Progression of Cardiovascular Calcification in End-Stage-Renal-Disease (ESRD) Patients on Hemodialysis (HD), NCT02966028.
Collapse
Affiliation(s)
- David A. Bushinsky
- Department of Medicine, University of Rochester School of Medicine, Rochester, New York
| | - Paolo Raggi
- Department of Medicine, Mazankowski Alberta Heart Institute and University of Alberta, Edmonton, Alberta, Canada
| | - Jordi Bover
- Department of Nephrology, Puigvert Foundation/Autonoma University, Sant Pau Biomedical Research Institute, Red de Investigacion Renal (REDinREN), Barcelona, Spain
| | - Markus Ketteler
- Department of General Internal Medicine and Nephrology, Robert Bosch Hospital, Stuttgart, Germany
| | - Antonio Bellasi
- Research, Innovation and Brand Reputation Unit, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Mariano Rodriguez
- Nephrology Unit, Reina Sofia University Hospital, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Red de Investigacion Renal (REDinREN), Córdoba, Spain
| | - Smeeta Sinha
- Department of Renal Medicine, Salford Royal National Health Service Foundation Trust, Salford, United Kingdom
| | - Rekha Garg
- Research and Development, Sanifit Therapeutics, San Diego, California
| | - Joan Perelló
- Research and Development, Sanifit Therapeutics, Palma, Spain,University Institute of Health Sciences Research (IUNICS-IDISBA), University of the Balearic Islands, Palma, Spain
| | - Alex Gold
- Research and Development, Sanifit Therapeutics, San Diego, California,Department of Medicine, Stanford University, Palo Alto, California
| | - Glenn M. Chertow
- Department of Medicine, Stanford University, Palo Alto, California
| | | | | |
Collapse
|
108
|
The effect of vitamin D supplementation on circulating osteoprogenitor cells: A pilot randomized controlled trial. Exp Gerontol 2021; 150:111399. [PMID: 33971278 DOI: 10.1016/j.exger.2021.111399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/27/2023]
Abstract
Circulating osteoprogenitor (COP) cells are a relatively newly discovered mesenchymal precursors population in the peripheral blood. While some aspects of their physiology have been documented in vitro, little is known about their behavior in vivo. To facilitate understanding regarding their potential role in the management of musculoskeletal disease, more research into how these cells respond to growth factors and hormones in vivo is still required. To this end, we performed a randomized controlled pilot study investigating the effect of vitamin D supplementation on COP cells in healthy older adults. Twenty-two individuals were recruited and stratified through their baseline vitamin D levels into deficient (<35 nmol/L), insufficient (35-49 nmol/L) and sufficient (>50 nmol/L) groups, and then randomized to receive either a 50,000 IU bolus dose of vitamin D, along with a 1000 IU daily supplement for six weeks, or the 1000 IU supplement alone. Participants were assessed at baseline, week three, and week six, with the primary outcome being a change in the number of COP cells. Secondary outcomes were vitamin D, markers of bone formation and resorption, parathyroid hormone, and calcium. The study showed that, independently of the dosing, increasing vitamin D levels led to a concomitant 52% increase in COP cell number (p < 0.001). There were no differences between strata, or any of the secondary outcomes in the trial. This suggests that COP cells are regulated in some way by vitamin D, similar to the bone marrow mesenchymal stem cell. Future studies are needed to evaluate the long-term effects of vitamin D supplementation, and how COP cells may be involved in chronic musculoskeletal disease.
Collapse
|
109
|
Dumortier C, Danopoulos S, Velard F, Al Alam D. Bone Cells Differentiation: How CFTR Mutations May Rule the Game of Stem Cells Commitment? Front Cell Dev Biol 2021; 9:611921. [PMID: 34026749 PMCID: PMC8139249 DOI: 10.3389/fcell.2021.611921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/12/2021] [Indexed: 12/30/2022] Open
Abstract
Cystic fibrosis (CF)-related bone disease has emerged as a significant comorbidity of CF and is characterized by decreased bone formation and increased bone resorption. Both osteoblast and osteoclast differentiations are impacted by cystic fibrosis transmembrane conductance regulator (CFTR) mutations. The defect of CFTR chloride channel or the loss of CFTRs ability to interact with other proteins affect several signaling pathways involved in stem cell differentiation and the commitment of these cells toward bone lineages. Specifically, TGF-, nuclear factor-kappa B (NF-B), PI3K/AKT, and MAPK/ERK signaling are disturbed by CFTR mutations, thus perturbing stem cell differentiation. High inflammation in patients changes myeloid lineage secretion, affecting both myeloid and mesenchymal differentiation. In osteoblast, Wnt signaling is impacted, resulting in consequences for both bone formation and resorption. Finally, CFTR could also have a direct role in osteoclasts resorptive function. In this review, we summarize the existing literature on the role of CFTR mutations on the commitment of induced pluripotent stem cells to bone cells.
Collapse
Affiliation(s)
- Claire Dumortier
- Division of Neonatology, Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States.,Universit de Reims Champagne-Ardenne, BIOS EA 4691, Reims, France
| | - Soula Danopoulos
- Division of Neonatology, Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Frdric Velard
- Universit de Reims Champagne-Ardenne, BIOS EA 4691, Reims, France
| | - Denise Al Alam
- Division of Neonatology, Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, United States
| |
Collapse
|
110
|
Duan L, Li Y, Hu J, Ma Q, Yu T, Zhang C, Luo F, Xu J, Dou C. Light rare earth elements hinder bone development via inhibiting type H vessels formation in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 218:112275. [PMID: 33962277 DOI: 10.1016/j.ecoenv.2021.112275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/14/2021] [Accepted: 04/18/2021] [Indexed: 06/12/2023]
Abstract
Light rare earth elements (LREEs) are widely used in medical, industrial, and agricultural fields. Wide application of light rare earth and exposure to these elements in human society leads to increasing accumulation of LREE in human skeletal system. However, the effects of LREEs on human bone health is not clear. In this study, we found that LREE reduced CD31highEmcnhigh endothelial cell mediated type H vessels formation at the metaphyseal sites, resulting in reduced bone mass and low bone quality in mouse bone development. To explore the underlying mechanism, we induced bone marrow macrophages (BMMs) to preosteoclasts (pOCs) with exposure of LREE (Pr3+, Nd3+, Sm3+). The cytotoxicity of LREE was evaluated by CCK-8. Platelet-derived growth factor (PDGF-BB) is the cytokine secreted by pOCs that most responsible for inducing Type H vessel formation. We used ELISA kit to determine the PDGF-BB level in pOC supernatant, and mouse serum finding that the PDGF-BB level was reduced by LREEs treatment. Then we tested the ability of migration and tube formation of HUVECs using condition medium from pOCs. The migration and tube formation ability of HUVECs were both suppressed with LREEs pretreatment. We concluded that LREEs hinder mouse bone development by suppressing type H vessels associated bone formation. DATA AND MATERIALS AVAILABILITY: All data generated or analyzed during this study are included in this article. Please contact the corresponding author for unique material requests. Some material used in the reported research may require requests to collaborators and agreements with both commercial and non-profit institutions, as specified in the paper. Requests are reviewed by Third Military Medical University to verify whether the request is subject to any intellectual property or confidentiality obligations. Any material that can be shared will be released via a Material Transfer Agreement.
Collapse
Affiliation(s)
- Lianli Duan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Yang Li
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Junxian Hu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Qinyu Ma
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Tao Yu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Chengmin Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China.
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
111
|
Liu N, Lu W, Qu X, Zhu C. LLLI promotes BMSC proliferation through circRNA_0001052/miR-124-3p. Lasers Med Sci 2021; 37:849-856. [PMID: 33884524 DOI: 10.1007/s10103-021-03322-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/11/2021] [Indexed: 12/13/2022]
Abstract
Osteoporosis (OP) is a multifactorial bone disease that occurs worldwide. The treatment of OP is still unsatisfactory. Bone mesenchymal stem cell (BMSC) differentiation is a key process in OP pathogenesis. Low-level laser irradiation (LLLI) has been reported to regulate BMSC proliferation, but the role of circRNAs in the LLLI-based promotion of BMSC proliferation remains unclear. CircRNAs are essential molecular regulators that participate in numerous biological processes and have therapeutic potential. miR-124-3p is an essential microRNA (miRNA), and its expression changes are related to BMSC proliferation ability. In the present study, gain-loss function of experiments demonstrated that circRNA_0001052 could regulate the proliferation of BMSCs by acting as a miR-124-3p sponge through the Wnt4/β-catenin pathway. The results of this study strongly suggest that circRNA_0001052 plays an essential role in BMSC proliferation in response to LLLI treatment, which is a potential therapeutic manipulation with clinical applications.
Collapse
Affiliation(s)
- Na Liu
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, Kunming, 650032, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.,Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
| | - Weiwei Lu
- Medical school, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xiaowen Qu
- Laser Medical Center, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 JinBi Road, Kunming, China.,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Chongtao Zhu
- Laser Medical Center, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 JinBi Road, Kunming, China. .,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China.
| |
Collapse
|
112
|
Novais A, Chatzopoulou E, Chaussain C, Gorin C. The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review. Cells 2021; 10:932. [PMID: 33920587 PMCID: PMC8073160 DOI: 10.3390/cells10040932] [Citation(s) in RCA: 27] [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: 03/05/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Bone is a hard-vascularized tissue, which renews itself continuously to adapt to the mechanical and metabolic demands of the body. The craniofacial area is prone to trauma and pathologies that often result in large bone damage, these leading to both aesthetic and functional complications for patients. The "gold standard" for treating these large defects is autologous bone grafting, which has some drawbacks including the requirement for a second surgical site with quantity of bone limitations, pain and other surgical complications. Indeed, tissue engineering combining a biomaterial with the appropriate cells and molecules of interest would allow a new therapeutic approach to treat large bone defects while avoiding complications associated with a second surgical site. This review first outlines the current knowledge of bone remodeling and the different signaling pathways involved seeking to improve our understanding of the roles of each to be able to stimulate or inhibit them. Secondly, it highlights the interesting characteristics of one growth factor in particular, FGF-2, and its role in bone homeostasis, before then analyzing its potential usefulness in craniofacial bone tissue engineering because of its proliferative, pro-angiogenic and pro-osteogenic effects depending on its spatial-temporal use, dose and mode of administration.
Collapse
Affiliation(s)
- Anita Novais
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Eirini Chatzopoulou
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
- Département de Parodontologie, Université de Paris, UFR Odontologie-Garancière, 75006 Paris, France
| | - Catherine Chaussain
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Caroline Gorin
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| |
Collapse
|
113
|
Barnsley J, Buckland G, Chan PE, Ong A, Ramos AS, Baxter M, Laskou F, Dennison EM, Cooper C, Patel HP. Pathophysiology and treatment of osteoporosis: challenges for clinical practice in older people. Aging Clin Exp Res 2021; 33:759-773. [PMID: 33742387 PMCID: PMC8084810 DOI: 10.1007/s40520-021-01817-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/13/2021] [Indexed: 12/12/2022]
Abstract
Osteoporosis, a common chronic metabolic bone disease is associated with considerable morbidity and mortality. As the prevalence of osteoporosis increases with age, a paralleled elevation in the rate of incident fragility fractures will be observed. This narrative review explores the origins of bone and considers physiological mechanisms involved in bone homeostasis relevant to management and treatment. Secondary causes of osteoporosis, as well as osteosarcopenia are discussed followed by an overview of the commonly used pharmacological treatments for osteoporosis in older people.
Collapse
Affiliation(s)
- J Barnsley
- Medicine for Older People, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - G Buckland
- Medicine for Older People, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - P E Chan
- Medicine for Older People, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - A Ong
- Medicine for Older People, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - A S Ramos
- Medicine for Older People, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - M Baxter
- Medicine for Older People, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - F Laskou
- MRC Lifecourse Epidemiology Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - E M Dennison
- MRC Lifecourse Epidemiology Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - C Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- University of Oxford, Oxford, UK
| | - Harnish P Patel
- Medicine for Older People, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
- MRC Lifecourse Epidemiology Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
- Academic Geriatric Medicine, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.
- NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and The University of Southampton, Southampton, UK.
| |
Collapse
|
114
|
Mechanical stimulation induced osteogenic differentiation of BMSCs through TWIST/E2A/p21 axis. Biosci Rep 2021; 40:222707. [PMID: 32309849 PMCID: PMC7199451 DOI: 10.1042/bsr20193876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/02/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
The relationship between mechanical force and alveolar bone remodeling is an important issue in orthodontics because tooth movement is dependent on the response of bone tissue to the mechanical force induced by the appliances used. Mechanical cyclical stretch plays an essential role in the cell osteogenic differentiation involved in bone remodeling. However, the underlying mechanisms are unclear, particularly the molecular pathways regulated by mechanical stimulation. In the present study, we reported a dynamic change of p21 level in response to mechanical cyclical stretch, and shRNA-p21 in bone marrow mesenchymal stem cells (BMSCs) induced osteogenic differentiation. The mechanism was mediated through TWIST/E2A/p21 axis. These results supported the mechanical stimulation-induced osteogenic differentiation is negatively regulated by p21.
Collapse
|
115
|
Yee MMF, Chin KY, Ima-Nirwana S, Wong SK. Vitamin A and Bone Health: A Review on Current Evidence. Molecules 2021; 26:1757. [PMID: 33801011 PMCID: PMC8003866 DOI: 10.3390/molecules26061757] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 11/16/2022] Open
Abstract
Vitamin A is a fat-soluble micronutrient essential for growth, immunity, and good vision. The preformed retinol is commonly found in food of animal origin whereas provitamin A is derived from food of plant origin. This review summarises the current evidence from animal, human and cell-culture studies on the effects of vitamin A towards bone health. Animal studies showed that the negative effects of retinol on the skeleton were observed at higher concentrations, especially on the cortical bone. In humans, the direct relationship between vitamin A and poor bone health was more pronounced in individuals with obesity or vitamin D deficiency. Mechanistically, vitamin A differentially influenced the stages of osteogenesis by enhancing early osteoblastic differentiation and inhibiting bone mineralisation via retinoic acid receptor (RAR) signalling and modulation of osteocyte/osteoblast-related bone peptides. However, adequate vitamin A intake through food or supplements was shown to maintain healthy bones. Meanwhile, provitamin A (carotene and β-cryptoxanthin) may also protect bone. In vitro evidence showed that carotene and β-cryptoxanthin may serve as precursors for retinoids, specifically all-trans-retinoic acid, which serve as ligand for RARs to promote osteogenesis and suppressed nuclear factor-kappa B activation to inhibit the differentiation and maturation of osteoclasts. In conclusion, we suggest that both vitamin A and provitamin A may be potential bone-protecting agents, and more studies are warranted to support this hypothesis.
Collapse
Affiliation(s)
| | | | | | - Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, University Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (M.M.F.Y.); (K.-Y.C.); (S.I.-N.)
| |
Collapse
|
116
|
Ziemińska M, Sieklucka B, Pawlak K. Vitamin K and D Supplementation and Bone Health in Chronic Kidney Disease-Apart or Together? Nutrients 2021; 13:809. [PMID: 33804453 PMCID: PMC7999920 DOI: 10.3390/nu13030809] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Vitamin K (VK) and vitamin D (VD) deficiency/insufficiency is a common feature of chronic kidney disease (CKD), leading to impaired bone quality and a higher risk of fractures. CKD patients, with disturbances in VK and VD metabolism, do not have sufficient levels of these vitamins for maintaining normal bone formation and mineralization. So far, there has been no consensus on what serum VK and VD levels can be considered sufficient in this particular population. Moreover, there are no clear guidelines how supplementation of these vitamins should be carried out in the course of CKD. Based on the existing results of preclinical studies and clinical evidence, this review intends to discuss the effect of VK and VD on bone remodeling in CKD. Although the mechanisms of action and the effects of these vitamins on bone are distinct, we try to find evidence for synergy between them in relation to bone metabolism, to answer the question of whether combined supplementation of VK and VD will be more beneficial for bone health in the CKD population than administering each of these vitamins separately.
Collapse
Affiliation(s)
- Marta Ziemińska
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Beata Sieklucka
- Department of Pharmacodynamics, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Krystyna Pawlak
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, 15-222 Bialystok, Poland;
| |
Collapse
|
117
|
Lo S, Fauzi MB. Current Update of Collagen Nanomaterials-Fabrication, Characterisation and Its Applications: A Review. Pharmaceutics 2021; 13:pharmaceutics13030316. [PMID: 33670973 PMCID: PMC7997363 DOI: 10.3390/pharmaceutics13030316] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 12/25/2022] Open
Abstract
Tissue engineering technology is a promising alternative approach for improvement in health management. Biomaterials play a major role, acting as a provisional bioscaffold for tissue repair and regeneration. Collagen a widely studied natural component largely present in the extracellular matrix (ECM) of the human body. It provides mechanical stability with suitable elasticity and strength to various tissues, including skin, bone, tendon, cornea and others. Even though exogenous collagen is commonly used in bioscaffolds, largely in the medical and pharmaceutical fields, nano collagen is a relatively new material involved in nanotechnology with a plethora of unexplored potential. Nano collagen is a form of collagen reduced to a nanoparticulate size, which has its advantages over the common three-dimensional (3D) collagen design, primarily due to its nano-size contributing to a higher surface area-to-volume ratio, aiding in withstanding large loads with minimal tension. It can be produced through different approaches including the electrospinning technique to produce nano collagen fibres resembling natural ECM. Nano collagen can be applied in various medical fields involving bioscaffold insertion or fillers for wound healing improvement; skin, bone, vascular grafting, nerve tissue and articular cartilage regeneration as well as aiding in drug delivery and incorporation for cosmetic purposes.
Collapse
|
118
|
Ramírez-Salazar EG, Almeraya EV, López-Perez TV, Patiño N, Salmeron J, Velázquez-Cruz R. MicroRNA-548-3p overexpression inhibits proliferation, migration and invasion in osteoblast-like cells by targeting STAT1 and MAFB. J Biochem 2021; 168:203-211. [PMID: 32196088 DOI: 10.1093/jb/mvaa033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoporosis is the most common bone disease and a public health issue with increasing prevalence in Mexico. This disease is caused by an imbalance in the bone remodelling process mediated by osteoclast and osteoblast. MicroRNAs have emerged as key players during the differentiation of both types of cells specialized involved in bone metabolism. We found high expression levels of miR-548x-3p in circulating monocytes derived from postmenopausal osteoporotic women. This study aimed to analyse the functional characterization of miR-548x-3p roles in the bone remodelling process. We validated by RT-qPCR, the elevated levels of miR-548x-3p in circulating monocytes derived from osteoporosis women. Through bioinformatics analysis, we identify MAFB and STAT1 as potential target genes for miR-548x-3p. Both genes showed low levels of expression in circulating monocytes derived from osteoporotic women. In addition, we demonstrated the binding of miR-548x-3p to the 3'-UTR of both mRNAs. MiR-548x-3p was overexpressed in osteoblasts-like cell lines decreasing the levels of MAFB and STAT1 mRNA and protein. We found that miR-548x-3p overexpression inhibits the proliferation, migration and invasion of the cell lines evaluated. Our results identified, by the first time, the potential role of miR-548x-3p as a modulator of the bone remodelling process by regulating the expression of MAFB and STAT1.
Collapse
Affiliation(s)
- Eric G Ramírez-Salazar
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
| | - Erika V Almeraya
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Tania V López-Perez
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Nelly Patiño
- Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
| | - Jorge Salmeron
- Centro de Investigación en Políticas, Población y Salud de la Facultad de Medicina-UNAM, Mexico City 04510, Mexico
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
119
|
Wu M, Liu J. Inhibitory effect of exogenous IL‐4 on orthodontic relapse in rats. Oral Dis 2021; 28:469-479. [DOI: 10.1111/odi.13763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/26/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Minting Wu
- Department of Prosthodontics School of Stomatology Jinan University Guangzhou510632China
- Center of Stomatology, The Second People's Hospital of Foshan (Affiliated Foshan Hospital of Southern Medical University) Foshan 528000 China
| | - Jing Liu
- Department of Prosthodontics School of Stomatology Jinan University Guangzhou510632China
| |
Collapse
|
120
|
Ekeuku SO, Pang KL, Chin KY. Effects of Caffeic Acid and Its Derivatives on Bone: A Systematic Review. Drug Des Devel Ther 2021; 15:259-275. [PMID: 33519191 PMCID: PMC7837552 DOI: 10.2147/dddt.s287280] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/18/2020] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Caffeic acid is a metabolite of hydroxycinnamate and phenylpropanoid, which are commonly synthesized by all plant species. It is present in various food sources that are known for their antioxidant properties. As an antioxidant, caffeic acid ameliorates reactive oxygen species, which have been reported to cause bone loss. Some studies have highlighted the effects of caffeic acid against bone resorption. METHODS A systematic review of the literature was conducted to identify relevant studies on the effects of caffeic acid on bone. A comprehensive search was conducted from July to November 2020 using PubMed, Scopus, Cochrane Library and Web of Science databases. Cellular, animal and human studies reporting the effects of caffeic acid, as a single compound, on bone cells or bone were considered. RESULTS The literature search found 226 articles on this topic, but only 24 articles met the inclusion criteria and were included in this review. The results showed that caffeic acid supplementation reduced osteoclastogenesis and bone resorption, possibly through its antioxidant potential and increased expression of osteoblast markers. However, some studies showed that caffeic acid did not affect bone resorption in ovariectomized rats and might impair bone mechanical properties in normal rats. CONCLUSION Caffeic acid potentially regulates the bone remodelling process by inhibiting osteoclastogenesis and bone resorption, as well as osteoblast apoptosis. Thus, it has medicinal values against bone diseases.
Collapse
Affiliation(s)
- Sophia Ogechi Ekeuku
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kok-Lun Pang
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
121
|
Zhu X, Chan YT, Yung PSH, Tuan RS, Jiang Y. Subchondral Bone Remodeling: A Therapeutic Target for Osteoarthritis. Front Cell Dev Biol 2021; 8:607764. [PMID: 33553146 PMCID: PMC7859330 DOI: 10.3389/fcell.2020.607764] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022] Open
Abstract
There is emerging awareness that subchondral bone remodeling plays an important role in the development of osteoarthritis (OA). This review presents recent investigations on the cellular and molecular mechanism of subchondral bone remodeling, and summarizes the current interventions and potential therapeutic targets related to OA subchondral bone remodeling. The first part of this review covers key cells and molecular mediators involved in subchondral bone remodeling (osteoclasts, osteoblasts, osteocytes, bone extracellular matrix, vascularization, nerve innervation, and related signaling pathways). The second part of this review describes candidate treatments for OA subchondral bone remodeling, including the use of bone-acting reagents and the application of regenerative therapies. Currently available clinical OA therapies and known responses in subchondral bone remodeling are summarized as a basis for the investigation of potential therapeutic mediators.
Collapse
Affiliation(s)
- Xiaobo Zhu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yau Tsz Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Patrick S H Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
122
|
Li H, Xiao Z, Quarles LD, Li W. Osteoporosis: Mechanism, Molecular Target and Current Status on Drug Development. Curr Med Chem 2021; 28:1489-1507. [PMID: 32223730 PMCID: PMC7665836 DOI: 10.2174/0929867327666200330142432] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/22/2022]
Abstract
CDATA[Osteoporosis is a pathological loss of bone mass due to an imbalance in bone remodeling where osteoclast-mediated bone resorption exceeds osteoblast-mediated bone formation resulting in skeletal fragility and fractures. Anti-resorptive agents, such as bisphosphonates and SERMs, and anabolic drugs that stimulate bone formation, including PTH analogues and sclerostin inhibitors, are current treatments for osteoporosis. Despite their efficacy, severe side effects and loss of potency may limit the long term usage of a single drug. Sequential and combinational use of current drugs, such as switching from an anabolic to an anti-resorptive agent, may provide an alternative approach. Moreover, there are novel drugs being developed against emerging new targets such as Cathepsin K and 17β-HSD2 that may have less side effects. This review will summarize the molecular mechanisms of osteoporosis, current drugs for osteoporosis treatment, and new drug development strategies.
Collapse
Affiliation(s)
- Hanxuan Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38165, USA
| | - L. Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38165, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| |
Collapse
|
123
|
Maia CDSF, Queiroz LY, de Oliveira IG, da Silva CCS, Cunha RA, Souza-Monteiro D, Ferreira MKM, Silveira FM, da Silva JC, Balbinot GDS, Collares FM, Martins MAT, Martins MD, Lima RR. Binge-Like Exposure During Adolescence Induces Detrimental Effects in Alveolar Bone that Persist in Adulthood. Alcohol Clin Exp Res 2021; 45:56-63. [PMID: 33165940 DOI: 10.1111/acer.14501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/28/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alcohol (EtOH) intake during adolescence has become an important public health issue. Although the detrimental effects of EtOH intake on the musculoskeletal system are well known, only a few studies have investigated its impact on the stomatognathic system of adolescents. This study aimed to investigate the effect of EtOH binge drinking on the alveolar bone and the long-term consequences after abstinence. METHODS Adolescent female Wistar rats (35 days old) were exposed to 4 cycles of EtOH binge drinking (3 g/kg/d; 3 days On-4 days Off) or distilled water (control group). Alveolar bone micromorphology and vertical bone distance were evaluated at 1, 30, and 60 days after that last EtOH intake through X-ray computed microtomography. The mineral:matrix ratio was assessed through Raman spectroscopy. RESULTS A decrease in both trabecular thickness and volume ratio, and an increase in trabecular separation were observed at the 1-day evaluation (immediate withdrawal). After 30 and 60 days, the alveolar bone parameters were found similar to control, except for the mineral:matrix ratio in the long-term abstinence. CONCLUSIONS EtOH binge drinking during adolescence results in alveolar bone damage that may persist in adulthood, even after abstinence.
Collapse
Affiliation(s)
- Cristiane do Socorro Ferraz Maia
- From the, Laboratory of Pharmacology of Inflammation and Behavior, (CdSFM, LYQ, IGdO, CCSdS), Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém, Pará, Brazil
| | - Letícia Yoshitome Queiroz
- From the, Laboratory of Pharmacology of Inflammation and Behavior, (CdSFM, LYQ, IGdO, CCSdS), Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém, Pará, Brazil
| | - Igor Gonçalves de Oliveira
- From the, Laboratory of Pharmacology of Inflammation and Behavior, (CdSFM, LYQ, IGdO, CCSdS), Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém, Pará, Brazil
| | - Carla Cristiane Soares da Silva
- From the, Laboratory of Pharmacology of Inflammation and Behavior, (CdSFM, LYQ, IGdO, CCSdS), Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém, Pará, Brazil
| | - Rodrigo A Cunha
- CNC-Center for Neurosciences and Cell Biology, (RAC), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Deiweson Souza-Monteiro
- Laboratory of Functional and Structural Biology, (DS-M, MKMF, RRL), Biological Science Institute, Federal University of Pará, Belém, Pará, Brazil
| | - Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, (DS-M, MKMF, RRL), Biological Science Institute, Federal University of Pará, Belém, Pará, Brazil
| | - Felipe Martins Silveira
- Department of Oral Pathology, (FMS, MDM), School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jordana Conceição da Silva
- Department of Dental Materials, (JCdS, GdSB, FMC), School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Gabriela de Souza Balbinot
- Department of Dental Materials, (JCdS, GdSB, FMC), School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabrício Mezzomo Collares
- Department of Dental Materials, (JCdS, GdSB, FMC), School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Marco Antônio Trevizani Martins
- Department of Oral Medcine, (MATM), Hospital de Clínicas de Porto Alegre (HCPA/UFRGS), University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Manoela Domingues Martins
- Department of Oral Pathology, (FMS, MDM), School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, (DS-M, MKMF, RRL), Biological Science Institute, Federal University of Pará, Belém, Pará, Brazil
| |
Collapse
|
124
|
Yang N, Liu Y. The Role of the Immune Microenvironment in Bone Regeneration. Int J Med Sci 2021; 18:3697-3707. [PMID: 34790042 PMCID: PMC8579305 DOI: 10.7150/ijms.61080] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/03/2021] [Indexed: 01/08/2023] Open
Abstract
Bone is an active tissue, being constantly renewed in healthy individuals with participation of the immune system to a large extent. Any imbalance between the processes of bone formation and bone resorption is linked to various inflammatory bone diseases. The immune system plays an important role in tissue formation and bone resorption. Recently, many studies have demonstrated complex interactions between the immune and skeletal systems. Both of immune cells and cytokines contribute to the regulation of bone homeostasis, and bone cells, including osteoblasts, osteoclasts, osteocytes, also influence the cellular functions of immune cells. These crosstalk mechanisms between the bone and immune system finally emerged, forming a new field of research called osteoimmunology. Therefore, the immune microenvironment is crucial in determining the speed and outcome of bone healing, repair, and regeneration. In this review, we summarise the role of the immune microenvironment in bone regeneration from the aspects of immune cells and immune cytokines. The elucidation of immune mechanisms involved in the process of bone regeneration would provide new therapeutic targets for improving the curative effects of bone injury treatment.
Collapse
Affiliation(s)
- Ning Yang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, China.,Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yao Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, Shenyang, China.,Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
125
|
Cheng L, Cai Z, Zhao J, Wang F, Lu M, Deng L, Cui W. Black phosphorus-based 2D materials for bone therapy. Bioact Mater 2020; 5:1026-1043. [PMID: 32695934 PMCID: PMC7355388 DOI: 10.1016/j.bioactmat.2020.06.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/22/2020] [Accepted: 06/08/2020] [Indexed: 02/08/2023] Open
Abstract
Since their discovery, Black Phosphorus (BP)-based nanomaterials have received extensive attentions in the fields of electromechanics, optics and biomedicine, due to their remarkable properties and excellent biocompatibility. The most essential feature of BP is that it is composed of a single phosphorus element, which has a high degree of homology with the inorganic components of natural bone, therefore it has a full advantage in the treatment of bone defects. This review will first introduce the source, physicochemical properties, and degradation products of BP, then introduce the remodeling process of bone, and comprehensively summarize the progress of BP-based materials for bone therapy in the form of hydrogels, polymer membranes, microspheres, and three-dimensional (3D) printed scaffolds. Finally, we discuss the challenges and prospects of BP-based implant materials in bone immune regulation and outlook the future clinical application.
Collapse
Affiliation(s)
- Liang Cheng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Zhengwei Cai
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, PR China
| | - Jingwen Zhao
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Fei Wang
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Min Lu
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, 1518 North Huancheng Road, Jiaxing 314000, PR China
| |
Collapse
|
126
|
Ma J, Zhu L, Zhou Z, Song T, Yang L, Yan X, Chen A, Ye TW. The calcium channel TRPV6 is a novel regulator of RANKL-induced osteoclastic differentiation and bone absorption activity through the IGF-PI3K-AKT pathway. Cell Prolif 2020; 54:e12955. [PMID: 33159483 PMCID: PMC7791174 DOI: 10.1111/cpr.12955] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Objectives Calcium ion signals are important for osteoclast differentiation. Transient receptor potential vanilloid 6 (TRPV6) is a regulator of bone homeostasis. However, it was unclear whether TRPV6 was involved in osteoclast formation. Therefore, the aim of this study was to evaluate the role of TPRV6 in bone metabolism and to clarify its regulatory role in osteoclasts at the cellular level. Materials and methods Bone structure and histological changes in Trpv6 knockout mice were examined using micro‐computed tomography and histological analyses. To investigate the effects of Trpv6 on osteoclast function, we silenced or overexpressed Trpv6 in osteoclasts via lentivirus transfection, respectively. Osteoclast differentiation and bone resorption viability were measured by tartrate‐resistant acid phosphatase (TRAP) staining and pit formation assays. The expression of osteoclast marker genes, including cathepsin k, DC‐STAMP, Atp6v0d2 and TRAP, was measured by qRT‐PCR. Cell immunofluorescence and Western blotting were applied to explore the mechanisms by which the IGF‐PI3K‐AKT pathway was involved in the regulation of osteoclast formation and bone resorption by Trpv6. Results We found that knockout of Trpv6 induced osteoporosis and enhanced bone resorption in mice, but did not affect bone formation. Further studies showed that Trpv6, which was distributed on the cell membrane of osteoclasts, acted as a negative regulator for osteoclast differentiation and function. Mechanistically, Trpv6 suppressed osteoclastogenesis by decreasing the ratios of phosphoprotein/total protein in the IGF–PI3K–AKT signalling pathway. Blocking of the IGF–PI3K–AKT pathway significantly alleviated the inhibitory effect of Trpv6 on osteoclasts formation. Conclusions Our study confirmed the important role of Trpv6 in bone metabolism and clarified its regulatory role in osteoclasts at the cellular level. Taken together, this study may inspire a new strategy for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Jun Ma
- Department of Orthopedic Trauma Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China.,Department of Health Statistics, The Second Military Medical University, Shanghai, China
| | - Lei Zhu
- Department of Orthopedic Trauma Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Zhibin Zhou
- Department of Orthopedic Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Tengfei Song
- Department of Orthopedic Trauma Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Lei Yang
- Department of Orthopedic Surgery, The 2nd affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xu Yan
- Department of Orthopedic Surgery, Naval Characteristic Medical Center, The Second Military Medical University, Shanghai, China
| | - Aimin Chen
- Department of Orthopedic Trauma Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Tian Wen Ye
- Department of Orthopedic Trauma Surgery, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
127
|
Li M, Xie Z, Li J, Lin J, Zheng G, Liu W, Tang S, Cen S, Ye G, Li Z, Yu W, Wang P, Wu Y, Shen H. GAS5 protects against osteoporosis by targeting UPF1/SMAD7 axis in osteoblast differentiation. eLife 2020; 9:e59079. [PMID: 33006314 PMCID: PMC7609060 DOI: 10.7554/elife.59079] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/30/2020] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis is a common systemic skeletal disorder resulting in bone fragility and increased fracture risk. It is still necessary to explore its detailed mechanisms and identify novel targets for the treatment of osteoporosis. Previously, we found that a lncRNA named GAS5 in human could negatively regulate the lipoblast/adipocyte differentiation. However, it is still unclear whether GAS5 affects osteoblast differentiation and whether GAS5 is associated with osteoporosis. Our current research found that GAS5 was decreased in the bones and BMSCs, a major origin of osteoblast, of osteoporosis patients. Mechanistically, GAS5 promotes the osteoblast differentiation by interacting with UPF1 to degrade SMAD7 mRNA. Moreover, a decreased bone mass and impaired bone repair ability were observed in Gas5 heterozygous mice, manifesting in osteoporosis. The systemic supplement of Gas5-overexpressing adenoviruses significantly ameliorated bone loss in an osteoporosis mouse model. In conclusion, GAS5 promotes osteoblast differentiation by targeting the UPF1/SMAD7 axis and protects against osteoporosis.
Collapse
Affiliation(s)
- Ming Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
| | - Jinteng Li
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| | - Jiajie Lin
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
| | - Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| | - Wenjie Liu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| | - Su'an Tang
- Department of Orthopedics, Zhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Shuizhong Cen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
- Department of Orthopedics, Zhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Guiwen Ye
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| | - Zhaofeng Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| | - Wenhui Yu
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
| | - Yanfeng Wu
- Center for Biotherapy,The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen UniversityShenzhenChina
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhouChina
| |
Collapse
|
128
|
Montaseri A, Giampietri C, Rossi M, Riccioli A, Fattore AD, Filippini A. The Role of Autophagy in Osteoclast Differentiation and Bone Resorption Function. Biomolecules 2020; 10:E1398. [PMID: 33008140 PMCID: PMC7601508 DOI: 10.3390/biom10101398] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022] Open
Abstract
Autophagy is an evolutionary conserved and highly regulated recycling process of cellular wastes. Having a housekeeping role, autophagy through the digestion of domestic cytosolic organelles, proteins, macromolecules, and pathogens, eliminates unnecessary materials and provides nutrients and energy for cell survival and maintenance. The critical role of autophagy and autophagy-related proteins in osteoclast differentiation, bone resorption, and maintenance of bone homeostasis has previously been reported. Increasing evidence reveals that autophagy dysregulation leads to alteration of osteoclast function and enhanced bone loss, which is associated with the onset and progression of osteoporosis. In this review, we briefly consolidate the current state-of-the-art technology regarding the role of autophagy in osteoclast function in both physiologic and pathologic conditions to have a more general view on this issue.
Collapse
Affiliation(s)
- Azadeh Montaseri
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (A.M.); (A.R.); (A.F.)
| | - Claudia Giampietri
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Human Anatomy, Sapienza University of Rome, 00161 Rome, Italy;
| | - Michela Rossi
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Anna Riccioli
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (A.M.); (A.R.); (A.F.)
| | - Andrea Del Fattore
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (A.M.); (A.R.); (A.F.)
| |
Collapse
|
129
|
Imtiyaz Z, Lin YT, Cheong UH, Jassey A, Liu HK, Lee MH. Compounds isolated from Euonymus spraguei Hayata induce ossification through multiple pathways. Saudi J Biol Sci 2020; 27:2227-2237. [PMID: 32884403 PMCID: PMC7451737 DOI: 10.1016/j.sjbs.2020.06.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 11/23/2022] Open
Abstract
The process of bone metabolism includes catabolism of old or mature bone and anabolism of new bone, carried out by osteoclasts and osteoblasts respectively. Any imbalance in this process results in loss of bone mass or osteoporosis. Drugs available to combat osteoporosis have certain adverse effects and are unable to improve bone formation, hence identifying new agents to fulfil these therapeutic gaps is required. To expand the scope of potential agents that enhance bone formation, we identified Euonymus spraguei Hayata as a plant material that possesses robust osteogenic potential using human osteoblast cells. We isolated three compounds, syringaresinol (1), syringin (2), and (−)-epicatechin (3), from E. spraguei. Results demonstrated that syringin (2), and (−)-epicatechin (3), increased alkaline phosphatase activity significantly up to 131.01% and 130.67%, respectively; they also elevated mineral deposition with respective values of up to 139.39% and 138.33%. In addition, 2 and 3 modulated autophagy and the bone morphogenetic protein (BMP)-2 signaling pathway. Our findings demonstrated that 2 and 3 induced osteogenesis by targeting multiple pathways and therefore can be considered as potent multi-targeted drugs for bone formation against osteoporosis.
Collapse
Affiliation(s)
- Zuha Imtiyaz
- Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Yi-Tzu Lin
- Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Ut-Hang Cheong
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Alagie Jassey
- College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Hui-Kang Liu
- Division of Basic Chinese Medicine, National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan
| | - Mei-Hsien Lee
- Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan.,Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan.,Center for Reproductive Medicine and Sciences, Taipei Medical University Hospital, Taipei 110, Taiwan
| |
Collapse
|
130
|
Fusion Potential of Human Osteoclasts In Vitro Reflects Age, Menopause, and In Vivo Bone Resorption Levels of Their Donors-A Possible Involvement of DC-STAMP. Int J Mol Sci 2020; 21:ijms21176368. [PMID: 32887359 PMCID: PMC7504560 DOI: 10.3390/ijms21176368] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/28/2022] Open
Abstract
It is well established that multinucleation is central for osteoclastic bone resorption. However, our knowledge on the mechanisms regulating how many nuclei an osteoclast will have is limited. The objective of this study was to investigate donor-related variations in the fusion potential of in vitro-generated osteoclasts. Therefore, CD14+ monocytes were isolated from 49 healthy female donors. Donor demographics were compared to the in vivo bone biomarker levels and their monocytes’ ability to differentiate into osteoclasts, showing that: (1) C-terminal telopeptide of type I collagen (CTX) and procollagen type I N-terminal propeptide (PINP) levels increase with age, (2) the number of nuclei per osteoclast in vitro increases with age, and (3) there is a positive correlation between the number of nuclei per osteoclast in vitro and CTX levels in vivo. Furthermore, the expression levels of the gene encoding dendritic cell-specific transmembrane protein (DCSTAMP) of osteoclasts in vitro correlated positively with the number of nuclei per osteoclast, CTX levels in vivo, and donor age. Our results furthermore suggest that these changes in gene expression may be mediated through age-related changes in DNA methylation levels. We conclude that both intrinsic factors and age-induced increase in fusion potential of osteoclasts could be contributing factors for the enhanced bone resorption in vivo, possibly caused by increased expression levels of DCSTAMP.
Collapse
|
131
|
Zhang V, Koa B, Borja AJ, Padmanhabhan S, Bhattaru A, Raynor WY, Rojulpote C, Seraj SM, Werner TJ, Rajapakse C, Alavi A, Revheim ME. Diagnosis and Monitoring of Osteoporosis with Total-Body 18F-Sodium Fluoride-PET/CT. PET Clin 2020; 15:487-496. [PMID: 32768370 DOI: 10.1016/j.cpet.2020.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In recent years, 18F-Sodium Fluoride (NaF)-PET/CT has seen its role in the detection and management of osteoporosis increase. This article reviews the extent of this application in the literature, its efficacy compared with other comparable imaging tools, and how total-body PET/CT combined with global disease assessment can revolutionize measurement of total osteoporotic disease activity. NaF-PET/CT eventually can be the modality of choice for metabolic bone disorders, especially with these advances in technology and computation.
Collapse
Affiliation(s)
- Vincent Zhang
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Koa
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Drexel University College of Medicine, Philadelphia, PA, USA
| | - Austin J Borja
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sayuri Padmanhabhan
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Abhijit Bhattaru
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - William Y Raynor
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Drexel University College of Medicine, Philadelphia, PA, USA
| | - Chaitanya Rojulpote
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Department of Internal Medicine, The Wright Center for Graduate Medical Education, Scranton, PA, USA
| | | | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Chamith Rajapakse
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Mona-Elisabeth Revheim
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
132
|
Tong L, Wang Y, Wang J, He F, Zhai J, Bai J, Zhu G. Radiation alters osteoclastogenesis by regulating the cytoskeleton and lytic enzymes in RAW 264.7 cells and mouse bone marrow-derived macrophages. Int J Radiat Biol 2020; 96:1296-1308. [PMID: 32687425 DOI: 10.1080/09553002.2020.1798542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE The aim of the present study was to investigate the duality of irradiation effect on osteoclastogenesis, particularly on the cytoskeleton and expression of lytic enzymes in osteoclast precursors. Therefore, the present study may serve as a useful reference for the prevention and treatment of radiation-induced bone loss in the clinic. MATERIALS AND METHODS Two typical osteoclast precursors, murine RAW 264.7 macrophage cells and mouse bone marrow-derived macrophages (BMMs), were exposed to radiation in the order of 0.25-8 Gy, and the effects on cell viability, TRAP activity and bone resorption were subsequently investigated. Furthermore, changes in the cytoskeleton, cell apoptosis, and expression of lytic enzymes in osteoclasts were examined to elucidate the molecular mechanism of the duality of irradiation on osteoclastogenesis. RESULTS Morphological changes and impaired viability were observed in RAW 264.7 cells and BMMs treated with 1-8 Gy irradiation with or without RANKL. However, the cell fusion tendency of osteoclasts was enhanced after 2 Gy irradiation, and an increased number of fused giant osteoclasts and enhanced F-actin ring formation were observed. Consistently, the bone resorption activity and the enzyme expression of TRAP, cathepsin K, matrix metalloproteinase 9, activator protein 1, and Caspase 9 were increased following irradiation with 2 Gy. Furthermore, intracellular ROS production and apoptosis of osteoclast precursors were increased. CONCLUSIONS Irradiation with 2 Gy inhibited the viability of osteoclast precursors, but increased osteoclastogenesis by enhancing cell fusion and increasing the secretion of lytic enzymes, which may be an important mechanism of radiation-induced bone loss.
Collapse
Affiliation(s)
- Ling Tong
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China.,Shanghai Municipal Center for Disease Control & Prevention, Shanghai, PR China
| | - Yuyang Wang
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Jianping Wang
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Feilong He
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China.,Shanghai Municipal Center for Disease Control & Prevention, Shanghai, PR China
| | - Jianglong Zhai
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Jiangtao Bai
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Guoying Zhu
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| |
Collapse
|
133
|
Møller AMJ, Delaissé JM, Olesen JB, Madsen JS, Canto LM, Bechmann T, Rogatto SR, Søe K. Aging and menopause reprogram osteoclast precursors for aggressive bone resorption. Bone Res 2020; 8:27. [PMID: 32637185 PMCID: PMC7329827 DOI: 10.1038/s41413-020-0102-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/06/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022] Open
Abstract
Women gradually lose bone from the age of ~35 years, but around menopause, the rate of bone loss escalates due to increasing bone resorption and decreasing bone formation levels, rendering these individuals more prone to developing osteoporosis. The increased osteoclast activity has been linked to a reduced estrogen level and other hormonal changes. However, it is unclear whether intrinsic changes in osteoclast precursors around menopause can also explain the increased osteoclast activity. Therefore, we set up a protocol in which CD14+ blood monocytes were isolated from 49 female donors (40-66 years old). Cells were differentiated into osteoclasts, and data on differentiation and resorption activity were collected. Using multiple linear regression analyses combining in vitro and in vivo data, we found the following: (1) age and menopausal status correlate with aggressive osteoclastic bone resorption in vitro; (2) the type I procollagen N-terminal propeptide level in vivo inversely correlates with osteoclast resorption activity in vitro; (3) the protein level of mature cathepsin K in osteoclasts in vitro increases with age and menopause; and (4) the promoter of the gene encoding the dendritic cell-specific transmembrane protein is less methylated with age. We conclude that monocytes are "reprogrammed" in vivo, allowing them to "remember" age, the menopausal status, and the bone formation status in vitro, resulting in more aggressive osteoclasts. Our discovery suggests that this may be mediated through DNA methylation. We suggest that this may have clinical implications and could contribute to understanding individual differences in age- and menopause-induced bone loss.
Collapse
Affiliation(s)
- Anaïs Marie Julie Møller
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Jean-Marie Delaissé
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
| | - Jacob Bastholm Olesen
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
| | - Jonna Skov Madsen
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Luisa Matos Canto
- Department of Clinical Genetics, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Troels Bechmann
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Silvia Regina Rogatto
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Genetics, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Kent Søe
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
- OPEN, Odense Patient data Explorative Network, Odense University Hospital, 5000 Odense C, Denmark
| |
Collapse
|
134
|
Bergamin LS, Penolazzi L, Lambertini E, Falzoni S, Sarti AC, Molle CM, Gendron FP, De Bonis P, Di Virgilio F, Piva R. Expression and function of the P2X7 receptor in human osteoblasts: The role of NFATc1 transcription factor. J Cell Physiol 2020; 236:641-652. [PMID: 32583512 DOI: 10.1002/jcp.29891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/21/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
Abstract
Bone mineralization is an orchestrated process by which mineral crystals are deposited by osteoblasts; however, the detailed mechanisms remain to be elucidated. The presence of P2X7 receptor (P2X7R) in immature and mature bone cells is well established, but contrasting evidence on its role in osteogenic differentiation and deposition of calcified bone matrix remains. To clarify these controversies in the present study, we investigated P2X7R participation in bone maturation. We demonstrated that the P2X7R is expressed and functional in human primary osteoblasts, and identified in the P2RX7 promoter several binding sites for transcription factors involved in bone mineralization. Of particular interest was the finding that P2X7R expression is enhanced by nuclear factor of activated T cells cytoplasmic 1 (NFATc1) overexpression, and accordingly, NFATc1 is recruited at the P2RX7 gene promoter in SaOS2 osteoblastic-like cells. In conclusion, our data provide further insights into the regulation of P2X7R expression and support the development of drugs targeting this receptor for the therapy of bone diseases.
Collapse
Affiliation(s)
| | - Letizia Penolazzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alba Clara Sarti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Caroline M Molle
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Fernand-Pierre Gendron
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Pasquale De Bonis
- Department of Neurosurgery, S. Anna University Hospital, Ferrara, Italy
| | | | - Roberta Piva
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
135
|
Abstract
Low levels of phosphate in blood secondary to a lack of phosphorous reabsorption in the renal tubule cause hypophosphataemic rickets. To increase blood calcium levels, bone calcium is released into the bloodstream by increasing the activity of osteoclasts in the bones. These alterations in bone metabolism lead to delayed growth and small height, diffuse bone pain, bone fragility with microfractures, and frequent fractures and bone deformities. Although management of some of these conditions is mainly conservative, in cases of bone deformities or severe osteomalacia surgery should be considered: from minimally invasive surgical techniques of guided growth in patients with less deformity and open physeal cartilage to correction osteotomies in three-dimensional deformities. This article briefly reviews the indications for surgery procedures to be performed in these patients, highlighting when to use aggressive and non-aggressive approaches.
Collapse
|
136
|
Mahmoud NS, Mohamed MR, Ali MAM, Aglan HA, Amr KS, Ahmed HH. Osteoblast-Based Therapy-A New Approach for Bone Repair in Osteoporosis: Pre-Clinical Setting. Tissue Eng Regen Med 2020; 17:363-373. [PMID: 32347454 DOI: 10.1007/s13770-020-00249-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Osteoporosis is a metabolic bone disease characterized by low bone density resulting in increased fracture susceptibility. This research was constructed to uncover the potential therapeutic application of osteoblasts transplantation, generated upon culturing male rat bone marrow-derived mesenchymal stem cells (BM-MSCs) in osteogenic medium (OM), OM containing gold (Au-NPs) or gold/hydroxyapatite (Au/HA-NPs) nanoparticles, in ovariectomized rats to counteract osteoporosis. METHODS Forty rats were randomized into: (1) negative control, (2) osteoporotic rats, whereas groups (3), (4) and (5) constituted osteoporotic rats treated with osteoblasts yielded from culturing BM-MSCs in OM, OM plus Au-NPs or Au/HA-NPs, respectively. After 3 months, osterix (OSX), bone alkaline phosphatase (BALP), sclerostin (SOST) and bone sialoprotein (BSP) serum levels were assessed. In addition, gene expression levels of cathepsin K, receptor activator of nuclear factor-κb ligand (RANKL), osteoprotegerin (OPG) and RANKL/OPG ratio were evaluated using real-time PCR. Moreover, histological investigation of femur bone tissues in different groups was performed. The homing of implanted osteoblasts to the osteoporotic femur bone of rats was documented by Sex determining region Y gene detection in bone tissue. RESULTS Our results indicated that osteoblasts infusion significantly blunted serum BALP, BSP and SOST levels, while significantly elevated OSX level. Also, they brought about significant down-regulation in gene expression levels of cathepsin K, RANKL and RANKL/OPG ratio versus untreated osteoporotic rats. Additionally, osteoblasts nidation could restore bone histoarchitecture. CONCLUSION These findings offer scientific evidence that transplanting osteoblasts in osteoporotic rats regains the homeostasis of the bone remodeling cycle, thus providing a promising treatment strategy for primary osteoporosis.
Collapse
Affiliation(s)
- Nadia Samy Mahmoud
- Hormones Department, Medical Research Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.)-Dokki, Giza, 12622, Egypt.
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.)-Dokki, Giza, 12622, Egypt.
| | - Mohamed Ragaa Mohamed
- Biochemistry Department, Faculty of Science, Ain Shams University, El-Khalyfa El-Ma'moun St., Abbasya, Cairo, 11566, Egypt
| | - Mohamed Ahmed Mohamed Ali
- Biochemistry Department, Faculty of Science, Ain Shams University, El-Khalyfa El-Ma'moun St., Abbasya, Cairo, 11566, Egypt
| | - Hadeer Ahmed Aglan
- Hormones Department, Medical Research Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.)-Dokki, Giza, 12622, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.)-Dokki, Giza, 12622, Egypt
| | - Khalda Sayed Amr
- Medical Molecular Genetics Department, Human Genetics and Genome Researches Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir St.)-Dokki, Giza, 12622, Egypt
| | - Hanaa Hamdy Ahmed
- Hormones Department, Medical Research Division, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.)-Dokki, Giza, 12622, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, 33 EL Bohouth St. (former EL -Tahrir st.)-Dokki, Giza, 12622, Egypt
| |
Collapse
|
137
|
Biological Factors, Metals, and Biomaterials Regulating Osteogenesis through Autophagy. Int J Mol Sci 2020; 21:ijms21082789. [PMID: 32316424 PMCID: PMC7215394 DOI: 10.3390/ijms21082789] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 01/18/2023] Open
Abstract
Bone loss raises great concern in numerous situations, such as ageing and many diseases and in both orthopedic and dentistry fields of application, with an extensive impact on health care. Therefore, it is crucial to understand the mechanisms and the determinants that can regulate osteogenesis and ensure bone balance. Autophagy is a well conserved lysosomal degradation pathway, which is known to be highly active during differentiation and development. This review provides a revision of the literature on all the exogen factors that can modulate osteogenesis through autophagy regulation. Metal ion exposition, mechanical stimuli, and biological factors, including hormones, nutrients, and metabolic conditions, were taken into consideration for their ability to tune osteogenic differentiation through autophagy. In addition, an exhaustive overview of biomaterials, both for orthopedic and dentistry applications, enhancing osteogenesis by modulation of the autophagic process is provided as well. Already investigated conditions regulating bone regeneration via autophagy need to be better understood for finely tailoring innovative therapeutic treatments and designing novel biomaterials.
Collapse
|
138
|
Chen K, Yan Z, Wang Y, Yang Y, Cai M, Huang C, Li B, Yang M, Zhou X, Wei X, Yang C, Chen Z, Zhai X, Li M. Shikonin mitigates ovariectomy-induced bone loss and RANKL-induced osteoclastogenesis via TRAF6-mediated signaling pathways. Biomed Pharmacother 2020; 126:110067. [PMID: 32272431 DOI: 10.1016/j.biopha.2020.110067] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Postmenopausal osteoporosis results from estrogen withdrawal and is characterized mainly by bone resorption. Shikonin is a bioactive constitute of Chinese traditional herb which plays a role in antimicrobial and antitumor activities. The study was designed to investigate the role of shikonin on postmenopausal osteoporosis and explore its underlying mechanisms. METHODS Immunofluorescence staining was performed to evaluate the effects of shikonin on actin ring formation. The expression levels of the nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) pathway were determined by Western blot analysis. To determine whether shikonin influences the receptor activator of nuclear factor-κB ligand (RANKL)-induced association between receptor activator of NF-κB (RANK) and tumor necrosis factor receptor associated factor 6 (TRAF6), immunofluorescence staining and immunoprecipitation experiments were performed. During our validation model, histomorphometric examination and micro-computed tomography (CT) were conducted to assess the morphology of osteoporosis. RESULTS Shikonin prevented bone loss by inhibiting osteoclastogenesis in vitro and improving bone loss in ovariectomized mice in vivo. At the molecular level, Western blot analysis indicated that shikonin inhibited the phosphorylation of inhibitor of NF-κB (IκB), P50, P65, extracellular regulated protein kinases (ERK), c-Jun N-terminal kinase (JNK), and P38. Interaction of TRAF6 and RANK was prevented, and downstream MAPK and NF-κB signaling pathways were downregulated. CONCLUSION Osteoclastic bone resorption was reduced in the presence of shikonin in vitro and in vivo. Shikonin is a promising candidate for treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Kai Chen
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Zijun Yan
- Graduate Management Unit, Shanghai Changhai Hospital, Navy Medical University, Shanghai 200433, China.
| | - Yiran Wang
- Shanghai Changhai Hospital, Navy Medical University, Shanghai 200433, China.
| | - Yilin Yang
- Shanghai Changhai Hospital, Navy Medical University, Shanghai 200433, China.
| | - Mengxi Cai
- Graduate Management Unit, Shanghai Changhai Hospital, Navy Medical University, Shanghai 200433, China.
| | - Chunyou Huang
- Graduate Management Unit, Shanghai Changhai Hospital, Navy Medical University, Shanghai 200433, China.
| | - Bo Li
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Mingyuan Yang
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Xiaoyi Zhou
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Xianzhao Wei
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Changwei Yang
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Ziqiang Chen
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Xiao Zhai
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| | - Ming Li
- Department of Orthopedics, Changhai Hospital of Navy Medical University, Shanghai 200433, China.
| |
Collapse
|
139
|
Guder C, Gravius S, Burger C, Wirtz DC, Schildberg FA. Osteoimmunology: A Current Update of the Interplay Between Bone and the Immune System. Front Immunol 2020; 11:58. [PMID: 32082321 PMCID: PMC7004969 DOI: 10.3389/fimmu.2020.00058] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Immunology, already a discipline in its own right, has become a major part of many different medical fields. However, its relationship to orthopedics and trauma surgery has unfortunately, and perhaps unjustly, been developing rather slowly. Discoveries in recent years have emphasized the immense breadth of communication and connection between both systems and, importantly, the highly promising therapeutic opportunities. Recent discoveries of factors originally assigned to the immune system have now also been shown to have a significant impact on bone health and disease, which has greatly changed how we approach treatment of bone pathologies. In case of bone fracture, immune cells, especially macrophages, are present throughout the whole healing process, assure defense against pathogens and discharge a complex variety of effectors to regulate bone modeling. In rheumatoid arthritis and osteoporosis, the immune system contributes to the formation of the pathological and chronic conditions. Fascinatingly, prosthesis failure is not at all solely a mechanical problem of improper strain but works in conjunction with an active contribution of the immune system as a reaction to irritant debris from material wear. Unraveling conjoined mechanisms of the immune and osseous systems heralds therapeutic possibilities for ailments of both. Contemplation of the bone as merely an unchanging support pillar is outdated and obsolete. Instead it is mandatory that this highly diverse network be incorporated in our understanding of the immune system and hematopoiesis.
Collapse
Affiliation(s)
- Christian Guder
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Sascha Gravius
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, University Medical Center Mannheim of University Heidelberg, Mannheim, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
140
|
Chai X, Zhang W, Chang B, Feng X, Song J, Li L, Yu C, Zhao J, Si H. GPR39 agonist TC-G 1008 promotes osteoblast differentiation and mineralization in MC3T3-E1 cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3569-3576. [PMID: 31448639 DOI: 10.1080/21691401.2019.1649270] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Osteoporosis-related bone fracture and falls have a severe impact on patients' daily lives. Osteoblasts are bone-building cells that play a vital role in bone formation and remodeling. Imbalanced osteoblast differentiation could lead to osteoporosis. GPR39 is an orphan G protein-coupled receptor that mediates metabolic pathways. In this study, we show that GPR39 is expressed in MC3T3-E1 cells. Osteoblast differentiation culture media induces GPR39, suggesting that GPR39 is a differentiation-responsive factor. Activation of GPR39 using its selective agonist TC-G 1008 induces alkaline phosphatase (ALP), osteocalcin (OCN), and type I collagen (Col-I) expression, and increases cellular ALP activity and calcium deposition, implying that GPR activation promotes cells toward osteoblast differentiation. Treatment with TC-G 1008 also increases Runx-2 expression and AMPK activation. However, the inhibition of AMPK by Compound C abolished TC-G 1008-mediated ALP, OCN, and Col-I induction, and reduces ALP activity and cellular calcium deposition as well as Runx-2 induction. These data indicate that TC-G 1008-mediated GPR39 activation involves AMPK-mediated Runx-2 induction. In summary, our study uncovers a new role of GPR39 activation in osteoblast differentiation, implying that GPR39 could be a promising therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Xingyu Chai
- School of Medicine, Shandong University , Ji'nan , China
| | - Wencan Zhang
- Department of Orthopedics, Qilu Hospital, Shandong University , Ji'nan , China
| | - Bingying Chang
- Department of Orthopedics, East Hospital of Shouguang People's Hospital , Weifang , China
| | - Xianli Feng
- Department of Orthopedics, Shandong Tai'an Coal Mine Hospital , Tai'an , China
| | - Jiang Song
- Department of Spine Surgery, Tengzhou Central People's Hospital , Tengzhou , China
| | - Le Li
- Department of Orthopedics, Qilu Hospital, Shandong University , Ji'nan , China
| | - Chenxiao Yu
- Department of Orthopedics, Qilu Hospital, Shandong University , Ji'nan , China
| | - Junyong Zhao
- College of Physics and Electronic Sciences, Shandong Normal University , Ji'nan , China
| | - Haipeng Si
- Department of Orthopedics, Qilu Hospital, Shandong University , Ji'nan , China
| |
Collapse
|
141
|
Shan SK, Lin X, Li F, Xu F, Zhong JY, Guo B, Wang Y, Zheng MH, Wu F, Yuan LQ. Exosomes and Bone Disease. Curr Pharm Des 2020; 25:4536-4549. [PMID: 31775592 DOI: 10.2174/1381612825666191127114054] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023]
Abstract
:
Exosomes, which mediate cell-to-cell communications and provide a novel insight into information
exchange, have drawn increasing attention in recent years. The homeostasis of bone metabolism is critical for
bone health. The most common bone diseases such as osteoporosis, osteoarthritis and bone fractures have apparent
correlations with exosomes. Accumulating evidence has suggested the potential regenerative capacities of
stem cell-derived exosomes. In this review, we summarise the pathophysiological mechanism, clinical picture and
therapeutic effects of exosomes in bone metabolism. We introduce the advantages and challenges in the application
of exosomes. Although the exact mechanisms remain unclear, miRNAs seem to play major roles in the
exosome.
Collapse
Affiliation(s)
- Su-Kang Shan
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao Lin
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Fuxingzi Li
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Xu
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Jia-Yu Zhong
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Bei Guo
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Wang
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Ming-Hui Zheng
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Wu
- Department of Pathology, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Ling-Qing Yuan
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Disease, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
142
|
Lalayiannis A, Crabtree N, Fewtrell M, Biassoni L, Milford D, Ferro C, Shroff R. Assessing bone mineralisation in children with chronic kidney disease: what clinical and research tools are available? Pediatr Nephrol 2020; 35:937-957. [PMID: 31240395 PMCID: PMC7184042 DOI: 10.1007/s00467-019-04271-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/19/2019] [Accepted: 04/26/2019] [Indexed: 12/11/2022]
Abstract
Mineral and bone disorder in chronic kidney disease (CKD-MBD) is a triad of biochemical imbalances of calcium, phosphate, parathyroid hormone and vitamin D, bone abnormalities and soft tissue calcification. Maintaining optimal bone health in children with CKD is important to prevent long-term complications, such as fractures, to optimise growth and possibly also to prevent extra-osseous calcification, especially vascular calcification. In this review, we discuss normal bone mineralisation, the pathophysiology of dysregulated homeostasis leading to mineralisation defects in CKD and its clinical consequences. Bone mineralisation is best assessed on bone histology and histomorphometry, but given the rarity with which this is performed, we present an overview of the tools available to clinicians to assess bone mineral density, including serum biomarkers and imaging such as dual-energy X-ray absorptiometry and peripheral quantitative computed tomography. We discuss key studies that have used these techniques, their advantages and disadvantages in childhood CKD and their relationship to biomarkers and bone histomorphometry. Finally, we present recommendations from relevant guidelines-Kidney Disease Improving Global Outcomes and the International Society of Clinical Densitometry-on the use of imaging, biomarkers and bone biopsy in assessing bone mineral density. Given low-level evidence from most paediatric studies, bone imaging and histology remain largely research tools, and current clinical management is guided by serum calcium, phosphate, PTH, vitamin D and alkaline phosphatase levels only.
Collapse
Affiliation(s)
- A.D. Lalayiannis
- Nephrology Department Great Ormond St. Hospital for Children NHS Foundation Trust and University College London Institute of Child Health, London, UK
| | - N.J. Crabtree
- Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | - M. Fewtrell
- Nephrology Department Great Ormond St. Hospital for Children NHS Foundation Trust and University College London Institute of Child Health, London, UK
| | - L. Biassoni
- Nephrology Department Great Ormond St. Hospital for Children NHS Foundation Trust and University College London Institute of Child Health, London, UK
| | - D.V. Milford
- Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | - C.J. Ferro
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - R. Shroff
- Nephrology Department Great Ormond St. Hospital for Children NHS Foundation Trust and University College London Institute of Child Health, London, UK
| |
Collapse
|
143
|
Gong X, Huang X, Yang Y, Zhou S, Dai Q, Jiang L. Local orthodontic force initiates widespread remodelling of the maxillary alveolar bone. AUSTRALASIAN ORTHODONTIC JOURNAL 2020. [DOI: 10.21307/aoj-2020-020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Abstract
Objectives
To clarify the effects of a local orthodontic force on alveolar bone by analysing bone remodelling in different regions of the maxilla during orthodontic tooth movement (OTM).
Methods
An OTM model was established in rats. Histological changes in the maxilla were analysed using TRAP staining, IHC staining for CTSK and haematoxylin and eosin (H and E) staining. The root bifurcation region of the alveolar bone of the first (M1), second (M2) and third (M3) molars were selected as the regions of interest (ROIs), which were further divided into a cervical and an apical level. Sequential fluorochrome labelling was performed to analyse bone deposition rates.
Results
The maxillary left first molars were moved mesially. TRAP staining and IHC staining for CTSK showed orthodontic force increased osteoclast numbers in all six ROIs at both the cervical and apical levels. H and E staining indicated elevated osteoblast numbers in the OTM group in all induced regions. Sequential fluorochrome labelling exhibited increased bone deposition rates around M1, M2 and M3 in the OTM group.
Conclusions
An orthodontic force applied to the first molar could initiate widespread remodelling of the maxillary alveolar bone, which was not restricted to the tension and pressure sites. This may revise the orthodontic biomechanical theory and provide new insights for clinical work.
Collapse
Affiliation(s)
- Xinyi Gong
- * Center of Craniofacial Orthodontics , Department of Oral and Cranio-maxillofacial Science , Ninth People’s Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China
- † The Affiliated Hospital of Stomatology , School of Stomatology , Zhejiang University School of Medicine , and University Key Laboratory of Oral Biomedical Research of Zhejiang Province , Hangzhou , P.R. China
| | - Xiangru Huang
- * Center of Craniofacial Orthodontics , Department of Oral and Cranio-maxillofacial Science , Ninth People’s Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China
| | - Yiling Yang
- * Center of Craniofacial Orthodontics , Department of Oral and Cranio-maxillofacial Science , Ninth People’s Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China
| | - Siru Zhou
- * Center of Craniofacial Orthodontics , Department of Oral and Cranio-maxillofacial Science , Ninth People’s Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China
| | - Qinggang Dai
- ± 2nd Dental Center , Ninth People’s Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China
| | - Lingyong Jiang
- * Center of Craniofacial Orthodontics , Department of Oral and Cranio-maxillofacial Science , Ninth People’s Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai , P.R. China
| |
Collapse
|
144
|
Isawa M, Karakawa A, Sakai N, Nishina S, Kuritani M, Chatani M, Negishi-Koga T, Sato M, Inoue M, Shimada Y, Takami M. Biological Effects of Anti-RANKL Antibody and Zoledronic Acid on Growth and Tooth Eruption in Growing Mice. Sci Rep 2019; 9:19895. [PMID: 31882595 PMCID: PMC6934544 DOI: 10.1038/s41598-019-56151-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/03/2019] [Indexed: 11/30/2022] Open
Abstract
The anti-bone resorptive drugs denosumab, an anti-human-RANKL antibody, and zoledronic acid (ZOL), a nitrogen-containing bisphosphonate, have recently been applied for treatment of pediatric patients with bone diseases, though details regarding their effects in growing children have yet to be fully elucidated. In the present study, we administered these anti-resorptive drugs to mice from the age of 1 week and continued once-weekly injections for a total of 7 times. Mice that received the anti-RANKL antibody displayed normal growth and tooth eruption, though osteopetrotic bone volume gain in long and alveolar bones was noted, while there were nearly no osteoclasts and a normal of number osteoblasts observed. In contrast, ZOL significantly delayed body growth, tooth root formation, and tooth eruption, with increased osteoclast and decreased osteoblast numbers. These findings suggest regulation of tooth eruption via osteoblast differentiation by some types of anti-resorptive drugs.
Collapse
Affiliation(s)
- Motoki Isawa
- Department of Pediatric Dentistry, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Akiko Karakawa
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Nobuhiro Sakai
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Saki Nishina
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Miku Kuritani
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
- Department of Special Needs Dentistry for Persons with Disabilities, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Masahiro Chatani
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Takako Negishi-Koga
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
- Department of Mucosal Barriology, International Research and Development for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Masashi Sato
- Department of Pediatric Dentistry, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Mitsuko Inoue
- Department of Pediatric Dentistry, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Yukie Shimada
- Department of Pediatric Dentistry, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Masamichi Takami
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan.
| |
Collapse
|
145
|
Fermented Oyster Extract Promotes Osteoblast Differentiation by Activating the Wnt/β-Catenin Signaling Pathway, Leading to Bone Formation. Biomolecules 2019; 9:biom9110711. [PMID: 31698882 PMCID: PMC6920898 DOI: 10.3390/biom9110711] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022] Open
Abstract
The Pacific oyster, Crassostrea gigas, is well-known as a nutritious food. Recently, we revealed that fermented extract of C. gigas (FO) inhibited ovariectomy-induced osteoporosis, resulting from suppression of osteoclastogenesis. However, since the beneficial effect of FO on osteogenesis is poorly understood, it was examined in mouse preosteoblast MC3T3-E1 cells, human osteosarcoma MG-63 osteoblast-like cells, and zebrafish larvae in this study. We found that FO increased mitochondrial activity from days 1 to 7; however, total cell number of MC3T3-E1 cells gradually decreased without any change in cell viability, which suggests that FO stimulates the differentiation of MC3T3-E1 cells. FO also promoted the expression of osteoblast marker genes, including runt-related transcription factor 2 (mRUNX2), alkaline phosphatase (mALP), collagen type I α1 (mCol1α1), osteocalcin (mOCN), osterix (mOSX), bone morphogenetic protein 2 (mBMP2), and mBMP4 in MC3T3-E1 cells accompanied by a significant increase in ALP activity. FO also increased nuclear translocation of RUNX2 and OSX transcription factors, ALP activity, and calcification in vitro along with the upregulated expression of osteoblast-specific marker proteins such as RUNX2, ALP, Col1α1, OCN, OSX, and BMP4. Additionally, FO enhanced bone mineralization (calcein intensity) in zebrafish larvae at 9 days post-fertilization comparable to that in the β-glycerophosphate (GP)-treated group. All the tested osteoblast marker genes, including zRUNX2a, zRUNX2b, zALP, zCol1a1, zOCN, zBMP2, and zBMP4, were also remarkably upregulated in the zebrafish larvae in response to FO. It also promoted tail fin regeneration in adult zebrafish as same as the GP-treated groups. Furthermore, not only FO positively regulate β-catenin expression and Wnt/β-catenin luciferase activity, but pretreatment with a Wnt/β-catenin inhibitor (FH535) also significantly decreased FO-mediated bone mineralization in zebrafish larvae, which indicates that FO-induced osteogenesis depends on the Wnt/β-catenin pathway. Altogether, the current study suggests that the supplemental intake of FO has a beneficial effect on osteogenesis.
Collapse
|
146
|
Lu J, Yang J, Zheng Y, Chen X, Fang S. Extracellular vesicles from endothelial progenitor cells prevent steroid-induced osteoporosis by suppressing the ferroptotic pathway in mouse osteoblasts based on bioinformatics evidence. Sci Rep 2019; 9:16130. [PMID: 31695092 PMCID: PMC6834614 DOI: 10.1038/s41598-019-52513-x] [Citation(s) in RCA: 45] [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: 04/29/2019] [Accepted: 10/17/2019] [Indexed: 12/21/2022] Open
Abstract
Abnormal antioxidative capabilities were observed in the pathogenesis of steroid-induced osteoporosis (SIOP). Ferroptosis is a recently discovered type of cell death that is characterized by the overproduction of ROS in response to GPX4 and system Xc- downregulation, which is mediated by an Fe2+ fenton reaction. However, investigations focusing on the relationship between ferroptosis and steroid-induced bone disease remain limited. In the present study, high-dose dexamethasone was used to establish a mouse SIOP model, and extracellular vesicles extracted from bone marrow-derived endothelial progenitor cells (EPC-EVs) alleviated the pathological changes in SIOP via microtomography (micro-CT), with elevations in bone volume (BV), bone surface (BS), trabecular thickness (Tb.Th), and trabecular connectivity density (Conn-D) and decreases in trabecular separation (Tb.sp) and the structure model index (SMI). Histopathological analysis, such as haematoxylin and eosin (HE) and Masson staining, showed that EPC-EVs treatment increased the volume and density of the trabecular bone and bone marrow. RNA sequencing (RNA-seq) and bioinformatics analysis revealed subcellular biological alterations upon steroid and EPC-EVs treatment. Compared with the control, high-dose dexamethasone downregulated GPX4 and system XC-, and the Kyoto Encyclopedia of Genes and Genomes (KEGG)-based gene set enrichment analysis suggested that the ferroptotic pathway was activated. In contrast, combination treatment with EPC-EVs partly reversed the KEGG-mapped changes in the ferroptotic pathway at both the gene and mRNA expression levels. In addition, alterations in ferroptotic marker expression, such as SLC3A2, SLC7A11, and GPX4, were further confirmed by RNA-seq. EPC-EVs were able to reverse dexamethasone treatment-induced alterations in cysteine and several oxidative injury markers, such as malondialdehyde (MDA), glutathione (GSH), and glutathione disulphide (GSSG) (as detected by ELISA). In conclusion, EPC-EVs prevented mouse glucocorticoid-induced osteoporosis by suppressing the ferroptotic pathway in osteoblasts, which may provide a basis for novel therapies for SIOP in humans.
Collapse
Affiliation(s)
- Jinsen Lu
- Department of Orthopaedics, Anhui Provincial Hospital, Anhui Medical University, Lujiang Road No. 17, 230001, Hefei, China.
| | - Jiazhao Yang
- Department of Orthopaedics, Anhui Provincial Hospital, Anhui Medical University, Lujiang Road No. 17, 230001, Hefei, China
| | - Yongshun Zheng
- Department of Orthopaedics, Anhui Provincial Hospital, Anhui Medical University, Lujiang Road No. 17, 230001, Hefei, China
| | - Xiaoyu Chen
- Department of Histology and Embryology, Anhui Medical University, Meishan Road No. 81, 230032, Hefei, China
| | - Shiyuan Fang
- Department of Orthopaedics, Anhui Provincial Hospital, Anhui Medical University, Lujiang Road No. 17, 230001, Hefei, China.
| |
Collapse
|
147
|
Corrigan MA, Coyle S, Eichholz KF, Riffault M, Lenehan B, Hoey DA. Aged Osteoporotic Bone Marrow Stromal Cells Demonstrate Defective Recruitment, Mechanosensitivity, and Matrix Deposition. Cells Tissues Organs 2019; 207:83-96. [PMID: 31655814 DOI: 10.1159/000503444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/18/2019] [Indexed: 11/19/2022] Open
Abstract
Bone formation requires the replenishment of the osteoblast from a progenitor or stem cell population, which must be recruited, expanded, and differentiated to ensure continued anabolism. How this occurs and whether it is altered in the osteoporotic environment is poorly understood. Furthermore, given that emerging treatments for osteoporosis are targeting this progenitor population, it is critical to determine the regenerative capacity of this cell type in the setting of osteoporosis. Human bone marrow stromal cells (hMSCs) from a cohort of aged osteoporotic patients were compared to MSCs isolated from healthy donors in terms of the ability to undergo recruitment and proliferation, and also respond to both the biophysical and biochemical cues that drive osteogenic matrix deposition. hMSCs isolated from healthy donors demonstrate good recruitment, mechanosensitivity, proliferation, and differentiation capacity. Contrastingly, hMSCs isolated from aged osteoporotic patients had significantly diminished regenerative potential. Interestingly, we demonstrated that osteoporotic hMSCs no longer responded to chemokine-directing recruitment and became desensitised to mechanical stimulation. The osteoporotic MSCs had a reduced proliferative potential and, importantly, they demonstrated an attenuated differentiation capability with reduced mineral and lipid formation. Moreover, during osteogenesis, despite minimal differences in the quantity of deposited collagen, the distribution of collagen was dramatically altered in osteoporosis, suggesting a potential defect in matrix quality. Taken together, this study has demonstrated that hMSCs isolated from aged osteoporotic patients demonstrate defective cell behaviour on multiple fronts, resulting in a significantly reduced regenerative potential, which must be considered during the development of new anabolic therapies that target this cell population.
Collapse
Affiliation(s)
- Michele A Corrigan
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Coyle
- Department of Trauma and Orthopaedics, University Hospital Limerick, Limerick, Ireland.,Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - Kian F Eichholz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Mathieu Riffault
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Brian Lenehan
- Department of Trauma and Orthopaedics, University Hospital Limerick, Limerick, Ireland.,Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - David A Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland, .,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland, .,Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin, Ireland,
| |
Collapse
|
148
|
Yorgan TA, Peters S, Amling M, Schinke T. Osteoblast-specific expression of Panx3 is dispensable for postnatal bone remodeling. Bone 2019; 127:155-163. [PMID: 31202927 DOI: 10.1016/j.bone.2019.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
Since cost-effective osteoanabolic treatment options remain to be established, it is relevant to identify specific molecules physiologically regulating osteoblast differentiation and/or activity that are principally accessible as drug targets. Specific or predominant gene expression in a given cell type often predicts a relevant function in the respective tissue. Thus, we aimed to identify genes encoding membrane-associated proteins with selective expression in differentiated osteoblasts. We therefore applied an unbiased approach, i.e. Affymetrix Gene Chip hybridization, to compare global gene expression in primary murine osteoblasts at two stages of differentiation. For the most strongly induced genes we analyzed their expression pattern in different tissues, which led us to identify known and unknown osteoblast differentiation markers with predominant expression in bone. One of these genes was Panx3, encoding a transmembrane hemichannel with ill-defined function in skeletal remodeling. To decipher the role of Panx3 in osteoblasts we first generated Panx3-fl/fl mice carrying a Runx2-Cre transgene. Using undecalcified histology followed by bone-specific histomorphometry we did not observe any significant difference between 24 weeks old Cre-negative and Cre-positive littermates. We additionally generated and analyzed mice with ubiquitous Panx3 deletion, where a delay of endochondral ossification did not translate into a detectable skeletal phenotype after weaning, possibly explained by compensatory induction of Panx1. Of note, newborn Panx3-deficient mice displayed significantly reduced serum glucose levels, which was not the case in older animals. Our findings demonstrate that Panx3 expression in osteoblasts is not required for postnatal bone remodeling, which essentially rules out its suitability as a target protein for osteoanabolic medication.
Collapse
Affiliation(s)
- Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany
| | - Stephanie Peters
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg 20246, Germany.
| |
Collapse
|
149
|
Glutamine Metabolism Is Essential for Stemness of Bone Marrow Mesenchymal Stem Cells and Bone Homeostasis. Stem Cells Int 2019; 2019:8928934. [PMID: 31611919 PMCID: PMC6757285 DOI: 10.1155/2019/8928934] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023] Open
Abstract
Skeleton has emerged as an endocrine organ which is both capable of regulating energy metabolism and being a target for it. Glutamine is the most bountiful and flexible amino acid in the body which provides adenosine 5′-triphosphate (ATP) demands for cells. Emerging evidences support that glutamine which acts as the second metabolic regulator after glucose exerts crucial roles in bone homeostasis at cellular level, including the lineage allocation and proliferation of bone mesenchymal stem cells (BMSCs), the matrix mineralization of osteoblasts, and the biosynthesis in chondrocytes. The integrated mechanism consisting of WNT, mammalian target of rapamycin (mTOR), and reactive oxygen species (ROS) signaling pathway in a glutamine-dependent pattern is responsible to regulate the complex intrinsic biological process, despite more extensive molecules are deserved to be elucidated in glutamine metabolism further. Indeed, dysfunctional glutamine metabolism enhances the development of degenerative bone diseases, such as osteoporosis and osteoarthritis, and glutamine or glutamine progenitor supplementation can partially restore bone defects which may promote treatment of bone diseases, although the mechanisms are not quite clear. In this review, we will summarize and update the latest research findings and clinical trials on the crucial regulatory roles of glutamine metabolism in BMSCs and BMSC-derived bone cells, also followed with the osteoclasts which are important in bone resorption.
Collapse
|
150
|
Jin Y, Long D, Li J, Yu R, Song Y, Fang J, Yang X, Zhou S, Huang S, Zhao Z. Extracellular vesicles in bone and tooth: A state-of-art paradigm in skeletal regeneration. J Cell Physiol 2019; 234:14838-14851. [PMID: 30847902 DOI: 10.1002/jcp.28303] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 02/05/2023]
Abstract
Bone and tooth, fundamental parts of the craniofacial skeleton, are anatomically and developmentally interconnected structures. Notably, pathological processes in these tissues underwent together and progressed in multilevels. Extracellular vesicles (EVs) are cell-released small organelles and transfer proteins and genetic information into cells and tissues. Although EVs have been identified in bone and tooth, particularly EVs have been identified in the bone formation and resorption, the concrete roles of EVs in bone and tooth development and diseases remain elusive. As such, we review the recent progress of EVs in bone and tooth to highlight the novel findings of EVs in cellular communication, tissue homeostasis, and interventions. This will enhance our comprehension on the skeletal biology and shed new light on the modulation of skeletal disorders and the potential of genetic treatment.
Collapse
Affiliation(s)
- Ying Jin
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China.,Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Juan Li
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Ruichao Yu
- Department of Pulmonary, Brigham and Women's Hospital, Harvard Medical School, Massachusetts
| | - Yueming Song
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Fang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Xi Yang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shu Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, P.R. China
| | - Shishu Huang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| |
Collapse
|