251
|
He X, Han Z, Jiang W, Huang F, Ren C, Wei Q, Zhou N. Hypoxia improved vasculogenesis in distraction osteogenesis through Mesenchymal-Epithelial transition (MET), Wnt/β-catenin signaling pathway, and autophagy. Acta Histochem 2020; 122:151593. [PMID: 32778247 DOI: 10.1016/j.acthis.2020.151593] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/16/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The osteogenesis rate of distraction osteogenesis is 4-6 times faster than that of infants, far beyond fracture healing. However, the osteogenesis mechanism of DO is complicated and inconclusive owing to two significant elements: mechanical tension which is well explored and trauma caused by bone fracture. Vasculogenesis and EPCs are critical for successful bone regeneration during DO. Thus, this study aimed to explore the effects of hypoxia caused by trauma or CoCl2 on the vasculogenesis of DO and EPCs. MATERIAL AND METHODS Mandibular DO and BF models were generated using 6 beagle dogs with a distraction rate of 1 mm per day for 7 days or acute lengthening for 7 mm. The vasculogenesis in DO-gap or BF-gap were assessed via histological analyses, qRT-PCR and immunofluorescence staining. Dog bone marrow EPCs were isolated and cultured with or without 0.1 mM CoCl2. The effect of hypoxia caused by CoCl2 were subsequently valuated via in vitro assays including Cell Counting Kit-8, transwell assay, qRT-PCR, western blot, and immunofluorescence staining. RESULTS Histological analyses, qRT-PCR and immunofluorescence staining revealed that vasculogenesis markedly accelerated in DO-gap compared with BF-gap, and the DO-gap displayed more positive to CD133, CD34, HIF-1α, E-cadherin, beclin1, β-catenin, VEGF, bFGF, and less positive to ZEB1 than BF-gap. In addition, in vitro analyses revealed CoCl2 treatment enhanced EPCs proliferation and migration, and the levels of HIF-1α, E-cadherin, β-catenin, beclin1, VEGF, bFGF of EPCs were increased, but the level of ZEB1 was decreased. CONCLUSION Our studies showed that hypoxia promoted vasculogenesis in DO and EPCs, and the mechanism may involve autophagy, Wnt/β-catenin signaling pathway, and Mesenchymal-Epithelial transition (MET).
Collapse
Affiliation(s)
- Xuan He
- College of Stomatology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, China; Guangxi Clinical Research Center for Craniofacial Deformity, China
| | - Zhiqi Han
- College of Stomatology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, China; Guangxi Clinical Research Center for Craniofacial Deformity, China
| | - Weidong Jiang
- College of Stomatology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, China; Guangxi Clinical Research Center for Craniofacial Deformity, China
| | - Fangfang Huang
- College of Stomatology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, China; Guangxi Clinical Research Center for Craniofacial Deformity, China
| | - Chao Ren
- College of Stomatology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, China; Guangxi Clinical Research Center for Craniofacial Deformity, China
| | - Qian Wei
- College of Stomatology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, China; Guangxi Clinical Research Center for Craniofacial Deformity, China
| | - Nuo Zhou
- College of Stomatology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, China; Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, China; Guangxi Clinical Research Center for Craniofacial Deformity, China.
| |
Collapse
|
252
|
Ruan Y, Kim HN, Ogana H, Kim YM. Wnt Signaling in Leukemia and Its Bone Marrow Microenvironment. Int J Mol Sci 2020; 21:ijms21176247. [PMID: 32872365 PMCID: PMC7503842 DOI: 10.3390/ijms21176247] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/16/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022] Open
Abstract
Leukemia is an aggressive hematologic neoplastic disease. Therapy-resistant leukemic stem cells (LSCs) may contribute to the relapse of the disease. LSCs are thought to be protected in the leukemia microenvironment, mainly consisting of mesenchymal stem/stromal cells (MSC), endothelial cells, and osteoblasts. Canonical and noncanonical Wnt pathways play a critical role in the maintenance of normal hematopoietic stem cells (HSC) and LSCs. In this review, we summarize recent findings on the role of Wnt signaling in leukemia and its microenvironment and provide information on the currently available strategies for targeting Wnt signaling.
Collapse
Affiliation(s)
- Yongsheng Ruan
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hye Na Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
| | - Heather Ogana
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
| | - Yong-Mi Kim
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90027, USA; (Y.R.); (H.N.K.); (H.O.)
- Correspondence:
| |
Collapse
|
253
|
Thorup AS, Dell'Accio F, Eldridge SE. Lessons from joint development for cartilage repair in the clinic. Dev Dyn 2020; 250:360-376. [PMID: 32738003 DOI: 10.1002/dvdy.228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.
Collapse
Affiliation(s)
- Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
254
|
Min Swe NM, Kobayashi Y, Kamimoto H, Moriyama K. Aberrantly activated Wnt/β-catenin pathway co-receptors LRP5 and LRP6 regulate osteoblast differentiation in the developing coronal sutures of an Apert syndrome (Fgfr2 S252W /+ ) mouse model. Dev Dyn 2020; 250:465-476. [PMID: 32822074 DOI: 10.1002/dvdy.239] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Apert syndrome is an autosomal, dominant inherited disorder characterized by craniosynostosis and syndactyly caused by gain-of-function mutations in the fibroblast growth factor receptor 2 (FGFR2) gene. Wnt/β-catenin signaling plays critical roles in regulating the skeletal development. Here, we analyzed the role of this pathway in the developing coronal sutures (CS) of a murine Apert syndrome model (Fgfr2S252W/+ ). RESULTS We observed aberrantly increased mRNA expression of Lrp5 and Lrp6 in CS of Fgfr2S252W/+ mice, whereas both wild type (WT) and Fgfr2S252W/+ mice showed similar expression of other Wnt/β-catenin-related genes, such as Wnt3, Wnt3a, Fzd4, Fzd6, Axin2, and Dkk1 as evidenced by in situ hybridization. Significantly increased Lrp5 and Lrp6 mRNA expression was observed by quantitative PCR analysis of cultured cells isolated from CS of Fgfr2S252W/+ mice. Phospho-LRP5, phospho-LRP6, and non-phospho-β-catenin were upregulated in Fgfr2S252W/+ CS compared with that in WT CS. Short-interfering RNA targeting Lrp5 and Lrp6 significantly reduced runt-related transcription factor 2, collagen type 1 alpha 1, and osteocalcin mRNA expression, and alkaline phosphatase activity in cultured cells. CONCLUSIONS The Wnt/β-catenin pathway was activated in the CS of Fgfr2S252W/+ mice during craniofacial development, suggesting the involvement of the Wnt/β-catenin pathway in the pathogenesis of CS synostosis in Fgfr2S252W/+ mice.
Collapse
Affiliation(s)
- Nay Myo Min Swe
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yukiho Kobayashi
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Kamimoto
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
255
|
On SW, Cho SW, Byun SH, Yang BE. Bioabsorbable Osteofixation Materials for Maxillofacial Bone Surgery: A Review on Polymers and Magnesium-Based Materials. Biomedicines 2020; 8:biomedicines8090300. [PMID: 32825692 PMCID: PMC7555479 DOI: 10.3390/biomedicines8090300] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 01/24/2023] Open
Abstract
Clinical application of osteofixation materials is essential in performing maxillofacial surgeries requiring rigid fixation of bone such as trauma surgery, orthognathic surgery, and skeletal reconstruction. In addition to the use of titanium plates and screws, clinical applications and attempts using bioabsorbable materials for osteofixation surgery are increasing with demands to avoid secondary surgery for the removal of plates and screws. Synthetic polymeric plates and screws were developed, reaching satisfactory physical properties comparable to those made with titanium. Although these polymeric materials are actively used in clinical practice, there remain some limitations to be improved. Due to questionable physical strength and cumbersome molding procedures, interests in resorbable metal materials for osteofixation emerged. Magnesium (Mg) gained attention again in the last decade as a new metallic alternative, and numerous animal studies to evaluate the possibility of clinical application of Mg-based materials are being conducted. Thanks to these researches and studies, vascular application of Mg-based biomaterials was successful; however, further studies are required for the clinical application of Mg-based biomaterials for osteofixation, especially in the facial skeleton. The review provides an overview of bioabsorbable osteofixation materials in maxillofacial bone surgery from polymer to Mg.
Collapse
Affiliation(s)
- Sung-Woon On
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Korea;
- Graduated School of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea; (S.-W.C.); (S.-H.B.)
- Institute of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea
| | - Seoung-Won Cho
- Graduated School of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea; (S.-W.C.); (S.-H.B.)
- Institute of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea
- Division of Oral and Maxillofacial Surgery, Hallym University Sacred Heart Hospital, Anyang 14066, Korea
| | - Soo-Hwan Byun
- Graduated School of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea; (S.-W.C.); (S.-H.B.)
- Institute of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea
- Division of Oral and Maxillofacial Surgery, Hallym University Sacred Heart Hospital, Anyang 14066, Korea
| | - Byoung-Eun Yang
- Graduated School of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea; (S.-W.C.); (S.-H.B.)
- Institute of Clinical Dentistry, Hallym University, Chuncheon 24252, Korea
- Division of Oral and Maxillofacial Surgery, Hallym University Sacred Heart Hospital, Anyang 14066, Korea
- Correspondence: ; Tel.: +82-380-3870
| |
Collapse
|
256
|
Ye G, Bao F, Zhang X, Song Z, Liao Y, Fei Y, Bunpetch V, Heng BC, Shen W, Liu H, Zhou J, Ouyang H. Nanomaterial-based scaffolds for bone tissue engineering and regeneration. Nanomedicine (Lond) 2020; 15:1995-2017. [PMID: 32812486 DOI: 10.2217/nnm-2020-0112] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The global incidence of bone tissue injuries has been increasing rapidly in recent years, making it imperative to develop suitable bone grafts for facilitating bone tissue regeneration. It has been demonstrated that nanomaterials/nanocomposites scaffolds can more effectively promote new bone tissue formation compared with micromaterials. This may be attributed to their nanoscaled structural and topological features that better mimic the physiological characteristics of natural bone tissue. In this review, we examined the current applications of various nanomaterial/nanocomposite scaffolds and different topological structures for bone tissue engineering, as well as the underlying mechanisms of regeneration. The potential risks and toxicity of nanomaterials will also be critically discussed. Finally, some considerations for the clinical applications of nanomaterials/nanocomposites scaffolds for bone tissue engineering are mentioned.
Collapse
Affiliation(s)
- Guo Ye
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Fangyuan Bao
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xianzhu Zhang
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Zhe Song
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Youguo Liao
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Yang Fei
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Varitsara Bunpetch
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing, PR China
| | - Weiliang Shen
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Hua Liu
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Jing Zhou
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Hongwei Ouyang
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| |
Collapse
|
257
|
Shu B, Zhao Y, Zhao S, Pan H, Xie R, Yi D, Lu K, Yang J, Xue C, Huang J, Wang J, Zhao D, Xiao G, Wang Y, Chen D. Inhibition of Axin1 in osteoblast precursor cells leads to defects in postnatal bone growth through suppressing osteoclast formation. Bone Res 2020; 8:31. [PMID: 32821442 PMCID: PMC7424530 DOI: 10.1038/s41413-020-0104-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022] Open
Abstract
Axin1 is a negative regulator of β-catenin signaling and its role in osteoblast precursor cells remains undefined. In the present studies, we determined changes in postnatal bone growth by deletion of Axin1 in osteoblast precursor cells and analyzed bone growth in newborn and postnatal Axin1Osx mice and found that hypertrophic cartilage area was largely expanded in Axin1Osx KO mice. A larger number of chondrocytes and unabsorbed cartilage matrix were found in the bone marrow cavity of Axin1Osx KO mice. Osteoclast formation in metaphyseal and subchondral bone areas was significantly decreased, demonstrated by decreased TRAP-positive cell numbers, associated with reduction of MMP9- and cathepsin K-positive cell numbers in Axin1Osx KO mice. OPG expression and the ratio of Opg to Rankl were significantly increased in osteoblasts of Axin1Osx KO mice. Osteoclast formation in primary bone marrow derived microphage (BMM) cells was significantly decreased when BMM cells were cultured with conditioned media (CM) collected from osteoblasts derived from Axin1Osx mice compared with BMM cells cultured with CM derived from WT mice. Thus, the loss of Axin1 in osteoblast precursor cells caused increased OPG and the decrease in osteoclast formation, leading to delayed bone growth in postnatal Axin1Osx KO mice.
Collapse
Affiliation(s)
- Bing Shu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Shitian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Haobo Pan
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 China
| | - Rong Xie
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Dan Yi
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 China
| | - Ke Lu
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Junjie Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Chunchun Xue
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Jian Huang
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Jing Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Dongfeng Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055 China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, 725 WanPing South Road, Shanghai, 200032 China
- Key Laboratory, Ministry of Education of China, 725 WanPing South Road, Shanghai, 200032 China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 China
| |
Collapse
|
258
|
Funato N. New Insights Into Cranial Synchondrosis Development: A Mini Review. Front Cell Dev Biol 2020; 8:706. [PMID: 32850826 PMCID: PMC7432265 DOI: 10.3389/fcell.2020.00706] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/13/2020] [Indexed: 11/24/2022] Open
Abstract
The synchondroses formed via endochondral ossification in the cranial base are an important growth center for the neurocranium. Abnormalities in the synchondroses affect cranial base elongation and the development of adjacent regions, including the craniofacial bones. In the central region of the cranial base, there are two synchondroses present—the intersphenoid synchondrosis and the spheno-occipital synchondrosis. These synchondroses consist of mirror image bipolar growth plates. The cross-talk of several signaling pathways, including the parathyroid hormone-like hormone (PTHLH)/parathyroid hormone-related protein (PTHrP), Indian hedgehog (Ihh), Wnt/β-catenin, and fibroblast growth factor (FGF) pathways, as well as regulation by cilium assembly and the transcription factors encoded by the RUNX2, SIX1, SIX2, SIX4, and TBX1 genes, play critical roles in synchondrosis development. Deletions or activation of these gene products in mice causes the abnormal ossification of cranial synchondrosis and skeletal elements. Gene disruption leads to both similar and markedly different abnormalities in the development of intersphenoid synchondrosis and spheno-occipital synchondrosis, as well as in the phenotypes of synchondroses and skeletal bones. This paper reviews the development of cranial synchondroses, along with its regulation by the signaling pathways and transcription factors, highlighting the differences between intersphenoid synchondrosis and spheno-occipital synchondrosis.
Collapse
Affiliation(s)
- Noriko Funato
- Department of Signal Gene Regulation, Tokyo Medical and Dental University, Tokyo, Japan.,Research Core, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
259
|
Ohsugi Y, Katagiri S, Hirota T, Niimi H, Hatasa M, Watanabe K, Shimohira T, Mizutani K, Kitazawa M, Matsuzawa A, Kadokura H, Yokose S, Iwata T, Aoki A. Laser irradiation decreases sclerostin expression in bone and osteogenic cells. FASEB J 2020; 34:12877-12893. [DOI: 10.1096/fj.202001032r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 01/25/2023]
Affiliation(s)
- Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Tomomitsu Hirota
- Division of Molecular Genetics, Research Center for Medical Science The Jikei University School of Medicine Tokyo Japan
| | - Hiromi Niimi
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Masahiro Hatasa
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Kazuki Watanabe
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Tsuyoshi Shimohira
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Koji Mizutani
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Moe Kitazawa
- Department of Epigenetics, Medical Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Ayumi Matsuzawa
- Department of Epigenetics, Medical Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Hiroshi Kadokura
- Division of Endodontic and Operative Dentistry, Department of Restorative and Biomaterials Sciences, School of Dentistry Meikai University Saitama Japan
| | - Satoshi Yokose
- Division of Endodontic and Operative Dentistry, Department of Restorative and Biomaterials Sciences, School of Dentistry Meikai University Saitama Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Akira Aoki
- Department of Periodontology, Graduate School of Medical and Dental Sciences Tokyo Medical and Dental University (TMDU) Tokyo Japan
| |
Collapse
|
260
|
Dash S, Trainor PA. The development, patterning and evolution of neural crest cell differentiation into cartilage and bone. Bone 2020; 137:115409. [PMID: 32417535 DOI: 10.1016/j.bone.2020.115409] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
Neural crest cells are a vertebrate-specific migratory, multipotent cell population that give rise to a diverse array of cells and tissues during development. Cranial neural crest cells, in particular, generate cartilage, bone, tendons and connective tissue in the head and face as well as neurons, glia and melanocytes. In this review, we focus on the chondrogenic and osteogenic potential of cranial neural crest cells and discuss the roles of Sox9, Runx2 and Msx1/2 transcription factors and WNT, FGF and TGFβ signaling pathways in regulating neural crest cell differentiation into cartilage and bone. We also describe cranioskeletal defects and disorders arising from gain or loss-of-function of genes that are required for patterning and differentiation of cranial neural crest cells. Finally, we discuss the evolution of skeletogenic potential in neural crest cells and their function as a conduit for intraspecies and interspecies variation, and the evolution of craniofacial novelties.
Collapse
Affiliation(s)
- Soma Dash
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
261
|
Oichi T, Otsuru S, Usami Y, Enomoto-Iwamoto M, Iwamoto M. Wnt signaling in chondroprogenitors during long bone development and growth. Bone 2020; 137:115368. [PMID: 32380258 PMCID: PMC7354209 DOI: 10.1016/j.bone.2020.115368] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 02/08/2023]
Abstract
Wnt signaling together with other signaling pathways governs cartilage development and the growth plate function during long bone formation and growth. β-catenin-dependent Wnt signaling is a specific lineage determinant of skeletal mesenchymal cells toward chondrogenic or osteogenic direction. Once cartilage forms and the growth plate organize, Wnt signaling continues to regulate proliferation and differentiation of the growth plate chondrocytes. Although chondrocytes in the growth plate have a high capacity to proliferate, new cells must be supplied to the growth plate from chondroprogenitor population. Advances in in vivo cell tracking techniques have demonstrated the importance of Wnt signaling in driving tissue renewal. The Wnt-responsive cells, genetically marked by the Wnt-reporter system, are found as stem cells in various tissues. Similarly, Wnt-responsive cells are found in the periphery of the growth plate and expanded to constitute entire column structure, indicating that Wnt signaling participates in the regulation of chondroprogenitors in the growth plate. This review will discuss advancements in research of progenitors in the growth plate, specifically focusing on Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Takeshi Oichi
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Satoru Otsuru
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Yu Usami
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Masahiro Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
262
|
Zhang J, He X, Chen X, Wu Y, Dong L, Cheng K, Lin J, Wang H, Weng W. Enhancing osteogenic differentiation of BMSCs on high magnetoelectric response films. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 113:110970. [DOI: 10.1016/j.msec.2020.110970] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/25/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022]
|
263
|
Ambrosi TH, Goodnough LH, Steininger HM, Hoover MY, Kim E, Koepke LS, Marecic O, Zhao L, Seita J, Bishop JA, Gardner MJ, Chan CKF. Geriatric fragility fractures are associated with a human skeletal stem cell defect. Aging Cell 2020; 19:e13164. [PMID: 32537886 PMCID: PMC7370785 DOI: 10.1111/acel.13164] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/24/2020] [Accepted: 04/19/2020] [Indexed: 12/22/2022] Open
Abstract
Fragility fractures have a limited capacity to regenerate, and impaired fracture healing is a leading cause of morbidity in the elderly. The recent identification of a highly purified bona fide human skeletal stem cell (hSSC) and its committed downstream progenitor cell populations provides an opportunity for understanding the mechanism of age‐related compromised fracture healing from the stem cell perspective. In this study, we tested whether hSSCs isolated from geriatric fractures demonstrate intrinsic functional defects that drive impaired healing. Using flow cytometry, we analyzed and isolated hSSCs from callus tissue of five different skeletal sites (n = 61) of patients ranging from 13 to 94 years of age for functional and molecular studies. We observed that fracture‐activated amplification of hSSC populations was comparable at all ages. However, functional analysis of isolated stem cells revealed that advanced age significantly correlated with reduced osteochondrogenic potential but was not associated with decreased in vitro clonogenicity. hSSCs derived from women displayed an exacerbated functional decline with age relative to those of aged men. Transcriptomic comparisons revealed downregulation of skeletogenic pathways such as WNT and upregulation of senescence‐related pathways in young versus older hSSCs. Strikingly, loss of Sirtuin1 expression played a major role in hSSC dysfunction but re‐activation by trans‐resveratrol or a small molecule compound restored in vitro differentiation potential. These are the first findings that characterize age‐related defects in purified hSSCs from geriatric fractures. Our results provide a foundation for future investigations into the mechanism and reversibility of skeletal stem cell aging in humans.
Collapse
Affiliation(s)
- Thomas H. Ambrosi
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| | - L. Henry Goodnough
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
- Department of Orthopaedic Surgery Stanford Hospital and Clinics Stanford CA USA
| | - Holly M. Steininger
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| | - Malachia Y. Hoover
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| | - Emiley Kim
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| | - Lauren S. Koepke
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| | - Owen Marecic
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| | - Liming Zhao
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| | - Jun Seita
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
- Medical Sciences Innovation Hub Program RIKEN Tokyo Japan
| | - Julius A. Bishop
- Department of Orthopaedic Surgery Stanford Hospital and Clinics Stanford CA USA
| | - Michael J. Gardner
- Department of Orthopaedic Surgery Stanford Hospital and Clinics Stanford CA USA
| | - Charles K. F. Chan
- Department of Surgery Stanford Medicine Stanford CA USA
- Institute for Stem Cell Biology and Regenerative Medicine Stanford Medicine Stanford CA USA
| |
Collapse
|
264
|
Kapania EM, Reif TJ, Tsumura A, Eby JM, Callaci JJ. Alcohol-induced Wnt signaling inhibition during bone fracture healing is normalized by intermittent parathyroid hormone treatment. Animal Model Exp Med 2020; 3:200-207. [PMID: 32613179 PMCID: PMC7323703 DOI: 10.1002/ame2.12116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 11/14/2022] Open
Abstract
Nearly half of orthopaedic trauma patients are intoxicated at the time of injury, and excess alcohol consumption increases the risk for fracture nonunion. Previous studies show alcohol disrupts fracture associated Wnt signaling required for normal bone fracture repair. Intermittent parathyroid hormone (PTH) promotes bone growth through canonical Wnt signaling, however, no studies have investigated the effect of PTH on alcohol-inhibited bone fracture repair. Male C57BL/6 mice received two-3 day alcohol binges separated by 4 days before receiving a mid-shaft tibia fracture. Postoperatively, mice received PTH daily until euthanasia. Wnt/β-catenin signaling was analyzed at 9 days post-fracture. As previously observed, acute alcohol exposure resulted in a >2-fold decrease in total and the active form of β-catenin and a 2-fold increase in inactive β-catenin within the fracture callus. Intermittent PTH abrogated the effect of alcohol on β-catenin within the fracture callus. Upstream of β-catenin, alcohol-treated animals had a 2-fold decrease in total LRP6, the Wnt co-receptor, which was restored with PTH treatment. Alcohol nor PTH had any significant effect on GSK-3β. These data show that intermittent PTH following a tibia fracture restores normal expression of Wnt signaling proteins within the fracture callus of alcohol-treated mice.
Collapse
Affiliation(s)
- Esha M. Kapania
- Internal Medicine‐Pediatric ResidentRush University Medical CenterChicagoILUSA
| | - Taylor J. Reif
- Limb Lengthening and Complex Reconstruction FellowshipHospital for Special SurgeryNew YorkNYUSA
| | - Aaron Tsumura
- Department of Orthopaedic Surgery and RehabilitationLoyola University Medical CenterMaywoodILUSA
| | - Jonathan M. Eby
- Department of Orthopaedic Surgery and RehabilitationLoyola University Medical CenterMaywoodILUSA
- Alcohol Research Program (ARP)Loyola University Chicago Stritch School of MedicineMaywoodILUSA
| | - John J. Callaci
- Department of Orthopaedic Surgery and RehabilitationLoyola University Medical CenterMaywoodILUSA
- Alcohol Research Program (ARP)Loyola University Chicago Stritch School of MedicineMaywoodILUSA
| |
Collapse
|
265
|
Iseri K, Dai L, Chen Z, Qureshi AR, Brismar TB, Stenvinkel P, Lindholm B. Bone mineral density and mortality in end-stage renal disease patients. Clin Kidney J 2020; 13:307-321. [PMID: 32699616 PMCID: PMC7367137 DOI: 10.1093/ckj/sfaa089] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis characterized by low bone mineral density (BMD) as assessed by dual-energy X-ray absorptiometry (DXA) is common among end-stage renal disease (ESRD) patients and associates with high fracture incidence and high all-cause mortality. This is because chronic kidney disease-mineral bone disorders (CKD-MBDs) promote not only bone disease (osteoporosis and renal dystrophy) but also vascular calcification and cardiovascular disease. The disturbed bone metabolism in ESRD leads to 'loss of cortical bone' with increased cortical porosity and thinning of cortical bone rather than to loss of trabecular bone. Low BMD, especially at cortical-rich bone sites, is closely linked to CKD-MBD, vascular calcification and poor cardiovascular outcomes. These effects appear to be largely mediated by shared mechanistic pathways via the 'bone-vascular axis' through which impaired bone status associates with changes in the vascular wall. Thus, bone is more than just the scaffolding that holds the body together and protects organs from external forces but is-in addition to its physical supportive function-also an active endocrine organ that interacts with the vasculature by paracrine and endocrine factors through pathways including Wnt signalling, osteoprotegerin (OPG)/receptor activator of nuclear factor-κB (RANK)/RANK ligand system and the Galectin-3/receptor of advanced glycation end products axis. The insight that osteogenesis and vascular calcification share many similarities-and the knowledge that vascular calcification is a cell-mediated active rather than a passive mineralization process-suggest that low BMD and vascular calcification ('vascular ossification') to a large extent represent two sides of the same coin. Here, we briefly review changes of BMD in ESRD as observed using different DXA methods (central and whole-body DXA) at different bone sites for BMD measurements, and summarize recent knowledge regarding the relationships between 'low BMD' and 'fracture incidence, vascular calcification and increased mortality' in ESRD patients, as well as potential 'molecular mechanisms' underlying these associations.
Collapse
Affiliation(s)
- Ken Iseri
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Division of Nephrology, Showa University School of Medicine, Tokyo, Japan
| | - Lu Dai
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Zhimin Chen
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Abdul Rashid Qureshi
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Torkel B Brismar
- Department of Clinical Science, Intervention and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Karolinska University Hospital, Huddinge, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Lindholm
- Department of Clinical Science, Intervention and Technology, Divisions of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
266
|
Hong H, Xu G, Deng H, Zhou X, Liu W, Cui Z. Concentration-Dependent Regulation of TiAl 6V 4 Particles on the Osteogenesis Potential of Human Bone Marrow Mesenchymal Stem Cells. Biol Trace Elem Res 2020; 195:445-453. [PMID: 31486015 DOI: 10.1007/s12011-019-01885-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Total joint replacement is one of the most effective treatments for osteoarthritis, while the aseptic loosening of artificial joint is a major complication leading to the joint replacement failure. There are very limited studies about the effects of titanium-alloy particles on the osteogenic differentiation of mesenchymal stem cells. In this study, human bone marrow-derived mesenchymal stem cells (BM-hMSCs) were treated with different concentrations of TiAl6V4 particles. The cell viability was detected by MTT assay, and the cell proliferation was assessed by CKK-8 assay. The early and late stages of osteogenic differentiation were determined by alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining assays. The expression of osteogenic genes and proteins was analyzed by RT-PCR and Western blot. TiAl6V4 particles at high concentration 100 μg/ml inhibited the cell viability of BM-hMSCs. However, TiAl6V4 in the range of 5-50 μg/ml did not show effects neither on the cell viability nor on the cell proliferation of BM-hMSCs. TiAl6V4 particles showed concentration-dependent bidirectional regulations on BM-hMSC osteogenesis. Specifically, TiAl6V4 at 5 μg/ml promoted the osteogenesis of BM-hMSCs, which was suppressed by TiAl6V4 at 50 μg/ml. Further, mechanism study revealed that the regulation of TiAl6V4 on BM-hMSCs was related to Wnt signaling pathway. Given the potential of mesenchymal cells, our study suggested that the minimization of metal use would be an attractive strategy to reduce the joint replacement failure.
Collapse
Affiliation(s)
- Hongxiang Hong
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Guanhua Xu
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Hongjian Deng
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Xiaogang Zhou
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Wei Liu
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China
| | - Zhiming Cui
- Department of Orthopaedics, the Second Affiliated Hospital of Nantong University, No. 6 North Hai-er-xiang Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
267
|
Sen B, Paradise CR, Xie Z, Sankaran J, Uzer G, Styner M, Meyer M, Dudakovic A, van Wijnen AJ, Rubin J. β-Catenin Preserves the Stem State of Murine Bone Marrow Stromal Cells Through Activation of EZH2. J Bone Miner Res 2020; 35:1149-1162. [PMID: 32022326 PMCID: PMC7295671 DOI: 10.1002/jbmr.3975] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 01/23/2020] [Accepted: 01/29/2020] [Indexed: 12/11/2022]
Abstract
During bone marrow stromal cell (BMSC) differentiation, both Wnt signaling and the development of a rigid cytoskeleton promote commitment to the osteoblastic over adipogenic lineage. β-catenin plays a critical role in the Wnt signaling pathway to facilitate downstream effects on gene expression. We show that β-catenin was additive with cytoskeletal signals to prevent adipogenesis, and β-catenin knockdown promoted adipogenesis even when the actin cytoskeleton was depolymerized. β-catenin also prevented osteoblast commitment in a cytoskeletal-independent manner, with β-catenin knockdown enhancing lineage commitment. Chromatin immunoprecipitation (ChIP)-sequencing demonstrated binding of β-catenin to the promoter of enhancer of zeste homolog 2 (EZH2), a key component of the polycomb repressive complex 2 (PRC2) complex that catalyzes histone methylation. Knockdown of β-catenin reduced EZH2 protein levels and decreased methylated histone 3 (H3K27me3) at osteogenic loci. Further, when EZH2 was inhibited, β-catenin's anti-differentiation effects were lost. These results indicate that regulating EZH2 activity is key to β-catenin's effects on BMSCs to preserve multipotentiality. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Buer Sen
- Department of Medicine, University of North Carolina Chapel Hill, Raleigh, NC, USA
| | - Christopher R Paradise
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zhihui Xie
- Department of Medicine, University of North Carolina Chapel Hill, Raleigh, NC, USA
| | - Jeyantt Sankaran
- Department of Medicine, University of North Carolina Chapel Hill, Raleigh, NC, USA
| | - Gunes Uzer
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Maya Styner
- Department of Medicine, University of North Carolina Chapel Hill, Raleigh, NC, USA
| | - Mark Meyer
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Janet Rubin
- Department of Medicine, University of North Carolina Chapel Hill, Raleigh, NC, USA
| |
Collapse
|
268
|
Xia T, Dong S, Tian J. miR‑29b promotes the osteogenic differentiation of mesenchymal stem cells derived from human adipose tissue via the PTEN/AKT/β‑catenin signaling pathway. Int J Mol Med 2020; 46:709-717. [PMID: 32468003 PMCID: PMC7307813 DOI: 10.3892/ijmm.2020.4615] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/10/2020] [Indexed: 01/02/2023] Open
Abstract
Accumulating evidence has documented that microRNAs (miRNAs or miRs) function as important post-transcriptional regulators of the differentiation of mesenchymal stem cells (MSCs), including human adipose-derived mesenchymal stem cells (hADSCs); however, their roles in hADSC osteogenic differentiation require further investigation. The present study aimed to investigate the role of miRNAs in the osteogenic differentiation of hADSCs and to elucidate the underlying molecular mechanisms. Using an miRNA microarray, it was found that 24 miRNAs were upregulated and 14 miRNAs were downregulated compared with the undifferentiated cells, and miR-29b-3p (miR-29b) was selected for further experiments. Functional experiments revealed that the upregulation of miR-29b by agomir-29b significantly enhanced alkaline phosphatase (ALP) activity and the mineralization of extracellular matrix (ECM), and led to an increase in the mRNA and protein levels of osteogenic marker genes, including runt-related transcription factor 2 (Runx2), osteopontin (OPN), osteocalcin (OCN) and bone sialoprotein (BSP), whereas the knockdown of miR-29b suppressed these processes. In addition, phosphatase and tensin homolog (PTEN), a negative regulator of the AKT/β-catenin pathway, was identified as a direct target of miR-29b in the hADSCs. Moreover, it was observed that the overexpression of miR-29b activated the AKT/β-catenin signaling pathway by inhibiting PTEN expression in the hADSCs. Most importantly, it was also found that the overexpression of PTEN reversed the promoting effects of miR-29b on osteogenic differentiation. On the whole, these findings suggest that miR-29b promotes the osteogenic differentiation of hADSCs by modulating the PTEN/AKT/β-catenin signaling pathway. Thus, this miRNA may be a promising target for the active modulation of hADSC-derived osteogenesis.
Collapse
Affiliation(s)
- Tian Xia
- Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| | - Shuanghai Dong
- Shanghai General Hospital of Nanjing Medical University, Shanghai 200080, P.R. China
| | - Jiwei Tian
- Department of Orthopedics, Shanghai Jiahui International Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
269
|
Pilquil C, Alvandi Z, Opas M. Calreticulin regulates a switch between osteoblast and chondrocyte lineages derived from murine embryonic stem cells. J Biol Chem 2020; 295:6861-6875. [PMID: 32220932 PMCID: PMC7242707 DOI: 10.1074/jbc.ra119.011029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 03/09/2020] [Indexed: 11/27/2022] Open
Abstract
Calreticulin is a highly conserved, ubiquitous Ca2+-buffering protein in the endoplasmic reticulum that controls transcriptional activity of various developmental programs and also of embryonic stem cell (ESC) differentiation. Calreticulin activates calcineurin, which dephosphorylates and induces the nuclear import of the osteogenic transcription regulator nuclear factor of activated T cells 1 (NFATC1). We investigated whether calreticulin controls a switch between osteogenesis and chondrogenesis in mouse ESCs through NFATC1. We found that in the absence of calreticulin, intranuclear transport of NFATC1 is blocked and that differentiation switches from osteogenic to chondrogenic, a process that could be mimicked by chemical inhibition of NFAT translocation. Glycogen synthase kinase 3β (GSK3β) deactivation and nuclear localization of β-catenin critical to osteogenesis were abrogated by calreticulin deficiency or NFAT blockade. Chemically induced GSK3β inhibition bypassed the calreticulin/calcineurin axis and increased osteoblast output from both control and calreticulin-deficient ESCs, while suppressing chondrogenesis. Calreticulin-deficient ESCs or cells treated with an NFAT blocker had enhanced expression of dickkopf WNT-signaling pathway inhibitor 1 (Dkk1), a canonical Wnt pathway antagonist that blocks GSK3β deactivation. The addition of recombinant mDKK1 switched osteogenic ESC differentiation toward chondrogenic differentiation. The results of our study indicate a role for endoplasmic reticulum calcium signaling via calreticulin in the differentiation of ESCs to closely associated osteoblast or chondrocyte lineages.
Collapse
Affiliation(s)
- Carlos Pilquil
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Zahra Alvandi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Michal Opas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
270
|
Evaluation of musculoskeletal phenotype of the G608G progeria mouse model with lonafarnib, pravastatin, and zoledronic acid as treatment groups. Proc Natl Acad Sci U S A 2020; 117:12029-12040. [PMID: 32404427 DOI: 10.1073/pnas.1906713117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a uniformly fatal condition that is especially prevalent in skin, cardiovascular, and musculoskeletal systems. A wide gap exists between our knowledge of the disease and a promising treatment or cure. The aim of this study was to first characterize the musculoskeletal phenotype of the homozygous G608G BAC-transgenic progeria mouse model, and to determine the phenotype changes of HGPS mice after a five-arm preclinical trial of different treatment combinations with lonafarnib, pravastatin, and zoledronic acid. Microcomputed tomography and CT-based rigidity analyses were performed to assess cortical and trabecular bone structure, density, and rigidity. Bones were loaded to failure with three-point bending to assess strength. Contrast-enhanced µCT imaging of mouse femurs was performed to measure glycosaminoglycan content, thickness, and volume of the femoral head articular cartilage. Advanced glycation end products were assessed with a fluorometric assay. The changes demonstrated in the cortical bone structure, rigidity, stiffness, and modulus of the HGPS G608G mouse model may increase the risk for bending and deformation, which could result in the skeletal dysplasia characteristic of HGPS. Cartilage abnormalities seen in this HGPS model resemble changes observed in the age-matched WT controls, including early loss of glycosaminoglycans, and decreased cartilage thickness and volume. Such changes might mimic prevalent degenerative joint diseases in the elderly. Lonafarnib monotherapy did not improve bone or cartilage parameters, but treatment combinations with pravastatin and zoledronic acid significantly improved bone structure and mechanical properties and cartilage structural parameters, which ameliorate the musculoskeletal phenotype of the disease.
Collapse
|
271
|
Kuriki M, Sato F, Arai HN, Sogabe M, Kaneko M, Kiyonari H, Kawakami K, Yoshimoto Y, Shukunami C, Sehara-Fujisawa A. Transient and lineage-restricted requirement of Ebf3 for sternum ossification. Development 2020; 147:147/9/dev186239. [PMID: 32398354 PMCID: PMC7240299 DOI: 10.1242/dev.186239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/20/2020] [Indexed: 12/18/2022]
Abstract
Osteoblasts arise from bone-surrounding connective tissue containing tenocytes and fibroblasts. Lineages of these cell populations and mechanisms of their differentiation are not well understood. Screening enhancer-trap lines of zebrafish allowed us to identify Ebf3 as a transcription factor marking tenocytes and connective tissue cells in skeletal muscle of embryos. Knockout of Ebf3 in mice had no effect on chondrogenesis but led to sternum ossification defects as a result of defective generation of Runx2+ pre-osteoblasts. Conditional and temporal Ebf3 knockout mice revealed requirements of Ebf3 in the lateral plate mesenchyme cells (LPMs), especially in tendon/muscle connective tissue cells, and a stage-specific Ebf3 requirement at embryonic day 9.5-10.5. Upregulated expression of connective tissue markers, such as Egr1/2 and Osr1, increased number of Islet1+ mesenchyme cells, and downregulation of gene expression of the Runx2 regulator Shox2 in Ebf3-deleted thoracic LPMs suggest crucial roles of Ebf3 in the onset of lateral plate mesoderm differentiation towards osteoblasts forming sternum tissues. Highlighted Article: Analysis of conditional and temporal knockout mice reveals roles of Ebf3 in the differentiation of lateral plate mesenchymal cells towards pre-osteoblasts during sternum ossification at the boundary of the lateral and somitic mesoderm.
Collapse
Affiliation(s)
- Mao Kuriki
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Fuminori Sato
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroyuki N Arai
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Maina Sogabe
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan.,Department of Muscle Aging and Regenerative Medicine, Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi, Tokyo 173-0015, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Atsuko Sehara-Fujisawa
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
272
|
Adipogenesis of skeletal muscle fibro/adipogenic progenitors is affected by the WNT5a/GSK3/β-catenin axis. Cell Death Differ 2020; 27:2921-2941. [PMID: 32382110 DOI: 10.1038/s41418-020-0551-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
Fibro/Adipogenic Progenitors (FAPs) are muscle-interstitial progenitors mediating pro-myogenic signals that are critical for muscle homeostasis and regeneration. In myopathies, the autocrine/paracrine constraints controlling FAP adipogenesis are released causing fat infiltrates. Here, by combining pharmacological screening, high-dimensional mass cytometry and in silico network modeling with the integration of single-cell/bulk RNA sequencing data, we highlighted the canonical WNT/GSK/β-catenin signaling as a crucial pathway modulating FAP adipogenesis triggered by insulin signaling. Consistently, pharmacological blockade of GSK3, by the LY2090314 inhibitor, stabilizes β-catenin and represses PPARγ expression abrogating FAP adipogenesis ex vivo while limiting fatty degeneration in vivo. Furthermore, GSK3 inhibition improves the FAP pro-myogenic role by efficiently stimulating, via follistatin secretion, muscle satellite cell (MuSC) differentiation into mature myotubes. Combining, publicly available single-cell RNAseq datasets, we characterize FAPs as the main source of WNT ligands inferring their potential in mediating autocrine/paracrine responses in the muscle niche. Lastly, we identify WNT5a, whose expression is impaired in dystrophic FAPs, as a crucial WNT ligand able to restrain the detrimental adipogenic differentiation drift of these cells through the positive modulation of the β-catenin signaling.
Collapse
|
273
|
Chen R, Hao Z, Chen X, Fu Q, Ma Y. Neuropeptide Y enhances proliferation and chondrogenic differentiation of ATDC5 cells. Neuropeptides 2020; 80:102022. [PMID: 31987472 DOI: 10.1016/j.npep.2020.102022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/19/2020] [Accepted: 01/19/2020] [Indexed: 12/19/2022]
Abstract
In recent years, emerging evidence has illustrated the indispensable role of sympathetic neurotransmitters and their receptors in cartilage mediation. The presence of neuropeptide Y (NPY)-positive sympathetic nerve fibres in cartilage and NPY-secretion function in chondrocytes raises the possibility of NPY directly regulating the function of chondrocytes. Therefore, this study intended to evaluate the effect of NPY and its receptors on the proliferation and chondrogenic differentiation of ATDC5 cells. Results showed NPY, especially at a concentration of 10-10 M, to significantly enhance proliferation of ATDC5 cells. Moreover, NPY effectively facilitated early chondrogenesis and late hypertrophy/mineralisation of ATDC5 cells via Y1 receptor signalling, rather than via Y2 receptor signalling. Taken together, the results help us to understand how NPY and its receptors affect the function of chondrocytes.
Collapse
Affiliation(s)
- Ruixin Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zhichao Hao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xiaodan Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Qiang Fu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| | - Yuanyuan Ma
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China.
| |
Collapse
|
274
|
Lotz EM, Cohen DJ, Schwartz Z, Boyan BD. Titanium implant surface properties enhance osseointegration in ovariectomy induced osteoporotic rats without pharmacologic intervention. Clin Oral Implants Res 2020; 31:374-387. [PMID: 31953969 PMCID: PMC7771214 DOI: 10.1111/clr.13575] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/03/2019] [Accepted: 01/04/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES This study determined whether implant surfaces that promote osseointegration in normal rats can promote osseointegration in osteoporotic rats without pharmacologic intervention. MATERIALS AND METHODS Virgin female 8-month-old CD Sprague Dawley rats (N = 25) were ovariectomized. At 6 weeks, microstructured/non-nanostructured/hydrophobic, microstructured/nanostructured/hydrophobic, or microstructured/nanostructured/hydrophilic Ti implants (Ø2.5 × 3.5 mm; Institut Straumann AG, Basel, Switzerland) were placed in the distal metaphysis of each femur. At 28 days, bone quality and implant osseointegration were assessed using microCT, histomorphometrics, and removal torque values (RTVs). Calvarial osteoblasts were isolated and cultured for 7 days on Ø15 mm Ti disks processed to exhibit similar surface characteristics as the implants used for the in vivo studies. The phenotype was assessed by measuring the production of osteocalcin, osteoprotegerin, osteopontin, BMP2, VEGF, and RANKL. RESULTS Microstructured/nanostructured/hydrophilic implants promoted increased bone-to-implant contact and RTVs in vivo and increased osteoblastic marker production in vitro compared to microstructured/non-nanostructured/hydrophobic and microstructured/nanostructured/hydrophobic implants, suggesting that osseointegration occurs in osteoporotic animals, and implant surface properties improve its rate. CONCLUSIONS Although all modified implants were able to osseointegrate in rats with OVX-induced osteoporosis without pharmacologic intervention, the degree of osseointegration was greater around microstructured/nanostructured/hydrophilic implant surfaces. These results suggest that when appropriate microstructure is present, hydrophilicity has a greater influence on Ti implant osseointegration compared to nanostructures. Moreover, modified implant surfaces can exert their control over the altered bone turnover observed in osteoporotic patients to stimulate functional osseointegration. These results provide critical insight for developing implants with improved osseointegration in patients with metabolic disorders of bone remodeling.
Collapse
Affiliation(s)
- Ethan M. Lotz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - David J. Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Barbara D. Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
275
|
Wang Y, Yang K, Li G, Liu R, Liu J, Li J, Tang M, Zhao M, Song J, Wen X. p75NTR -/- mice exhibit an alveolar bone loss phenotype and inhibited PI3K/Akt/β-catenin pathway. Cell Prolif 2020; 53:e12800. [PMID: 32215984 PMCID: PMC7162804 DOI: 10.1111/cpr.12800] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/18/2020] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The aim of this study was to investigate the role of p75 neurotrophin receptor (p75NTR) in regulating the mouse alveolar bone development and the mineralization potential of murine ectomesenchymal stem cells (EMSCs). Moreover, we tried to explore the underlying mechanisms associated with the PI3K/Akt/β-catenin pathway. MATERIALS AND METHODS p75NTR knockout (p75NTR-/- ) mice and wild-type (WT) littermates were used. E12.5d p75NTR-/- and WT EMSCs were isolated in the same pregnant p75NTR-/+ mice from embryonic maxillofacial processes separately. Mouse alveolar bone mass was evaluated using micro-CT. Differential osteogenic differentiation pathways between p75NTR-/- and WT EMSCs were analysed by RNA-sequencing. The PI3K inhibitor LY294002 and PI3K agonist 740Y-P were used to regulate the PI3K/Akt pathway in EMSCs. p75NTR overexpression lentiviruses, p75NTR knock-down lentiviruses and recombined mouse NGF were used to transfect cells. RESULTS The alveolar bone mass was found reduced in the p75NTR knockout mouse comparing to the WT mouse. During mineralization induction, p75NTR-/- EMSCs displayed decreased osteogenic capacity and downregulated PI3K/Akt/β-catenin signalling. The PI3K/Akt/β-catenin pathway positively regulates the potential of differential mineralization in EMSCs. The promotive effect of p75NTR overexpression can be attenuated by LY294002, while the inhibitory effect of p75NTR knock-down on Runx2 and Col1 expression can be reversed by 740Y-P. CONCLUSION Deletion of p75NTR reduced alveolar bone mass in mice. P75NTR positively regulated the osteogenic differentiation of EMSCs via enhancing the PI3K/Akt/β-catenin pathway.
Collapse
Affiliation(s)
- Yingying Wang
- Department of StomatologyDaping HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Kun Yang
- Department of PeriodontologyStomatological HospitalZunyi Medical UniversityZunyiChina
| | - Gang Li
- Department of StomatologyDaping HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Rui Liu
- Department of StomatologyDaping HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Junyu Liu
- College of StomatologyChongqing Medical UniversityChongqingChina
| | - Jun Li
- Department of StomatologyDaping HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Mengying Tang
- Hospital of StomatologySouthwest Medical UniversityLuzhouChina
| | - Manzhu Zhao
- College of StomatologyChongqing Medical UniversityChongqingChina
| | - Jinlin Song
- College of StomatologyChongqing Medical UniversityChongqingChina
| | - Xiujie Wen
- Department of StomatologyDaping HospitalArmy Medical University (Third Military Medical University)ChongqingChina
- Hospital of StomatologySouthwest Medical UniversityLuzhouChina
| |
Collapse
|
276
|
Kaji DA, Tan Z, Johnson GL, Huang W, Vasquez K, Lehoczky JA, Levi B, Cheah KS, Huang AH. Cellular Plasticity in Musculoskeletal Development, Regeneration, and Disease. J Orthop Res 2020; 38:708-718. [PMID: 31721278 PMCID: PMC7213644 DOI: 10.1002/jor.24523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 02/04/2023]
Abstract
In this review, we highlight themes from a recent workshop focused on "Plasticity of Cell Fate in Musculoskeletal Tissues" held at the Orthopaedic Research Society's 2019 annual meeting. Experts in the field provided examples of mesenchymal cell plasticity during normal musculoskeletal development, regeneration, and disease. A thorough understanding of the biology underpinning mesenchymal cell plasticity may offer a roadmap for promoting regeneration while attenuating pathologic differentiation. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:708-718, 2020.
Collapse
Affiliation(s)
- Deepak A. Kaji
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, NYC, NY, USA
| | - Zhijia Tan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Gemma L. Johnson
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, USA,Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Wesley Huang
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kaetlin Vasquez
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jessica A. Lehoczky
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Benjamin Levi
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Alice H. Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, NYC, NY, USA
| |
Collapse
|
277
|
Liu Y, Fang J, Zhang Q, Zhang X, Cao Y, Chen W, Shao Z, Yang S, Wu D, Hung M, Zhang Y, Tong W, Tian H. Wnt10b-overexpressing umbilical cord mesenchymal stem cells promote critical size rat calvarial defect healing by enhanced osteogenesis and VEGF-mediated angiogenesis. J Orthop Translat 2020; 23:29-37. [PMID: 32477867 PMCID: PMC7248289 DOI: 10.1016/j.jot.2020.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/08/2020] [Accepted: 02/13/2020] [Indexed: 01/04/2023] Open
Abstract
Background/objectives Accelerating the process of bone regeneration is of great interest for surgeons and basic scientists alike. Recently, umbilical cord mesenchymal stem cells (UCMSCs) are considered clinically applicable for tissue regeneration due to their noninvasive harvesting and better viability. Nonetheless, the bone regenerative ability of human UCMSCs (HUCMSCs) is largely unknown. This study aimed to investigate whether Wnt10b-overexpressing HUCMSCs have enhanced bone regeneration ability in a rat model. Method A rat calvarial defect was performed on 8-week old male Sprague Dawley rats. Commercially purchased HUCMSCsEmp in hydrogel, HUCMSCsWnt10b in hydrogel and HUCMSCsWnt10b with IWR-1 were placed in the calvarial bone defect right after surgery on rats (N = 8 rats for each group). Calvaria were harvested for micro-CT analysis and histology four weeks after surgery. CFU-F and multi-differentiation assay by oil red staining, alizarin red staining and RT-PCR (real-time polymerase chain reaction) were performed on HUCMSCsEmp and HUCMSCsWnt10bin vitro. Conditioned media from HUCMSCsEmp and HUCMSCsWnt10b were collected and used to treat human umbilical cord vein endothelial cells in Matrigel to access vessel formation capacity by tube formation assay. Results Alizarin red staining, oil red staining and RT-PCR results showed robust osteogenic differentiation but poor adipogenic differentiation ability of HUCMSCsWnt10b. Furthermore, HUCMSCsWnt10b could accelerate bone defect healing, which was likely due to enhanced angiogenesis after the HUCMSCsWnt10b treatment, because more CD31+ vessels and increased vascular endothelial growth factor-A (VEGF-A) expression were observed, compared with the HUCMSCsEmp treatment. Conditioned media from HUCMSCsWnt10b also induced endothelial cells to form vessel tubes in a tube formation assay, which could be abolished by SU5416, an angiogenesis inhibitor. Conclusion To our knowledge, this is the first study providing empirical evidence that HUCMSCsWnt10b can enhance their ability to heal calvarial bone defects via VEGF-mediated angiogenesis. The translational potential of this article HUCMSCsWnt10b can accelerate critical size calvaria and are a new promising therapeutic cell source for fracture nonunion healing.
Collapse
Affiliation(s)
- Yong Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Jiarui Fang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Quan Zhang
- Wuhan Hamilton Biotechnology Co., Ltd, Wuhan, Hubei, 430075, China
| | - Xiaoguang Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Yulin Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Wei Chen
- The Third Hospital of Hebei Medical University, 139, Ziqiang Road, Shi Jiazhuang, Hebei, 050051, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Shuhua Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Dongcheng Wu
- Wuhan Hamilton Biotechnology Co., Ltd, Wuhan, Hubei, 430075, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, China
| | - Man Hung
- College of Dental Medicine, Roseman University of Health Sciences, 10984 S River Front Pkwy, South Jordan, UT, 84095, USA
| | - Yingze Zhang
- The Third Hospital of Hebei Medical University, 139, Ziqiang Road, Shi Jiazhuang, Hebei, 050051, China
| | - Wei Tong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Hongtao Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| |
Collapse
|
278
|
Shi L, Feng L, Zhu ML, Yang ZM, Wu TY, Xu J, Liu Y, Lin WP, Lo JHT, Zhang JF, Li G. Vasoactive Intestinal Peptide Stimulates Bone Marrow-Mesenchymal Stem Cells Osteogenesis Differentiation by Activating Wnt/β-Catenin Signaling Pathway and Promotes Rat Skull Defect Repair. Stem Cells Dev 2020; 29:655-666. [PMID: 32070222 DOI: 10.1089/scd.2019.0148] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bone defect regeneration is a complex process that involves the coordination of a variety of different type of cells. As bone tissues are innervated and rich in nerve fibers, the neuropeptides released from various never fibers could regulate bone development, metabolism, and remodeling. Among all the neuropeptides, vasoactive intestinal peptide (VIP) could modulate the functions of both osteoblasts and osteoclasts, and may play a vital role in bone marrow mesenchymal stem cell (BMSC) osteogenesis during bone repair. In this study, we investigated the role of VIP in bone formation and the mechanisms of VIP in mediating BMSC osteogenic differentiation, and its possibility in clinical application of bone defect reconstruction. Our in vitro study results indicated that VIP promoted BMSC osteogenic differentiation by activating Wnt/β-catenin signaling pathway in BMSCs. VIP could also stimulate tube formation of EA.hy926 endothelial cell and increase vascular endothelial growth factor (VEGF) expression in BMSCs. Furthermore, in the rat skull defect model, VIP-conjugated functionalized hydrogel significantly enhanced cranial bone defect repair compared with the control group, with increased bone formation and angiogenesis. Taken together, as a member of neuropeptides, VIP could promote the BMSCs osteogenesis and angiogenesis differentiation in vitro and stimulate bone repair in vivo by activating Wnt/β-catenin signaling pathway. The knowledge obtained from this study emphasized the close association between innervation and bone repair process, and VIP may be a potential therapeutic agent for augmenting bone repair.
Collapse
Affiliation(s)
- Liu Shi
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China.,Trauma Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China.,School of Medicine, Southeast University, Nanjing, P.R. China
| | - Lu Feng
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China
| | - Mei-Ling Zhu
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China
| | - Zheng-Meng Yang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China
| | - Tian-Yi Wu
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China.,Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Jia Xu
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China
| | - Yang Liu
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China
| | - Wei-Ping Lin
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China
| | - Jessica Hiu Tung Lo
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China
| | - Jin-Fang Zhang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China.,Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, P.R. China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, P.R. China
| |
Collapse
|
279
|
Zhou T, Gao B, Fan Y, Liu Y, Feng S, Cong Q, Zhang X, Zhou Y, Yadav PS, Lin J, Wu N, Zhao L, Huang D, Zhou S, Su P, Yang Y. Piezo1/2 mediate mechanotransduction essential for bone formation through concerted activation of NFAT-YAP1-ß-catenin. eLife 2020; 9:52779. [PMID: 32186512 PMCID: PMC7112954 DOI: 10.7554/elife.52779] [Citation(s) in RCA: 210] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/17/2020] [Indexed: 12/15/2022] Open
Abstract
Mechanical forces are fundamental regulators of cell behaviors. However, molecular regulation of mechanotransduction remain poorly understood. Here, we identified the mechanosensitive channels Piezo1 and Piezo2 as key force sensors required for bone development and osteoblast differentiation. Loss of Piezo1, or more severely Piezo1/2, in mesenchymal or osteoblast progenitor cells, led to multiple spontaneous bone fractures in newborn mice due to inhibition of osteoblast differentiation and increased bone resorption. In addition, loss of Piezo1/2 rendered resistant to further bone loss caused by unloading in both bone development and homeostasis. Mechanistically, Piezo1/2 relayed fluid shear stress and extracellular matrix stiffness signals to activate Ca2+ influx to stimulate Calcineurin, which promotes concerted activation of NFATc1, YAP1 and ß-catenin transcription factors by inducing their dephosphorylation as well as NFAT/YAP1/ß-catenin complex formation. Yap1 and ß-catenin activities were reduced in the Piezo1 and Piezo1/2 mutant bones and such defects were partially rescued by enhanced ß-catenin activities.
Collapse
Affiliation(s)
- Taifeng Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Bo Gao
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Fan
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Shuhao Feng
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangdong, China
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Xiaolei Zhang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yaxing Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Prem S Yadav
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| | - Jiachen Lin
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States.,Department of Orthopedic Surgery and Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Nan Wu
- Department of Orthopedic Surgery and Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Liang Zhao
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangdong, China
| | - Dongsheng Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, United States
| | - Peiqiang Su
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, United States
| |
Collapse
|
280
|
Qin X, Jiang Q, Miyazaki T, Komori T. Runx2 regulates cranial suture closure by inducing hedgehog, Fgf, Wnt and Pthlh signaling pathway gene expressions in suture mesenchymal cells. Hum Mol Genet 2020; 28:896-911. [PMID: 30445456 DOI: 10.1093/hmg/ddy386] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 10/25/2018] [Accepted: 11/02/2018] [Indexed: 12/23/2022] Open
Abstract
Cleidocranial dysplasia (CCD, #119600), which is characterized by hypoplastic clavicles, open fontanelles, supernumerary teeth and a short stature, is caused by heterozygous mutations in RUNX2. However, it currently remains unclear why suture closure is severely impaired in CCD patients. The closure of posterior frontal (PF) and sagittal (SAG) sutures was completely interrupted in Runx2+/- mice, and the proliferation of suture mesenchymal cells and their condensation were less than those in wild-type mice. To elucidate the underlying molecular mechanisms, differentially expressed genes between wild-type and Runx2+/- PF and SAG sutures were identified by microarray and real-time reverse transcription polymerase chain reaction analyses. The expression of hedgehog, Fgf, Wnt and Pthlh signaling pathway genes, including Gli1, Ptch1, Ihh, Fgfr2, Fgfr3, Tcf7, Wnt10b and Pth1r, which were directly regulated by Runx2, was reduced in the sutures, but not the calvarial bone tissues of Runx2+/- mice. Bone formation and suture closure were enhanced in an organ culture of Runx2+/- calvariae with ligands or agonists of hedgehog, Fgf, Wnt and Pthlh signaling, while they were suppressed and suture mesenchymal cell proliferation was decreased in an organ culture of wild-type calvariae with their antagonists. These results indicate that more than a half dosage of Runx2 is required for the proliferation of suture mesenchymal cells, their condensation and commitment to osteoblast-lineage cells, and the induction of hedgehog, Fgf, Wnt and Pthlh signaling pathway gene expressions in sutures, but not in calvarial bone tissues, and also that the activation of hedgehog, Fgf, Wnt and Pthlh signaling pathways is necessary for suture closure.
Collapse
Affiliation(s)
- Xin Qin
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Qing Jiang
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toshihiro Miyazaki
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
281
|
Zhao X, Xie L, Wang Z, Wang J, Xu H, Han X, Bai D, Deng P. ZBP1 (DAI/DLM-1) promotes osteogenic differentiation while inhibiting adipogenic differentiation in mesenchymal stem cells through a positive feedback loop of Wnt/β-catenin signaling. Bone Res 2020; 8:12. [PMID: 32195010 PMCID: PMC7058036 DOI: 10.1038/s41413-020-0085-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/19/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023] Open
Abstract
The lineage specification of mesenchymal stem/stromal cells (MSCs) is tightly regulated by a wide range of factors. Recently, the versatile functions of ZBP1 (also known as DAI or DLM-1) have been reported in the blood circulation and immune systems. However, the biological function of ZBP1 during the lineage specification of MSCs is still unknown. In the present study, we found that ZBP1 was upregulated during osteogenesis but downregulated during adipogenesis in mouse bone marrow-derived MSCs (mBMSCs). ZBP1 was highly expressed in osteoblasts but expressed at a relatively low level in marrow adipocytes. Knockdown of ZBP1 inhibited alkaline phosphataseactivity, extracellular matrix mineralization, and osteogenesis-related gene expression in vitro and reduced ectopic bone formation in vivo. Knockdown of ZBP1 also promoted adipogenesis in MSCs in vitro. Conversely, the overexpression of ZBP1 increased the osteogenesis but suppressed the adipogenesis of MSCs. When the expression of ZBP1 was rescued, the osteogenic capacity of ZBP1-depleted mBMSCs was restored at both the molecular and phenotypic levels. Furthermore, we demonstrated that ZBP1, a newly identified target of Wnt/β-catenin signaling, was required for β-catenin translocation into nuclei. Collectively, our results indicate that ZBP1 is a novel regulator of bone and fat transdifferentiation via Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xuefeng Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Zhiyong Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Hao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Peng Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| |
Collapse
|
282
|
Kim BJ, Lee SH, Koh JM. Potential Biomarkers to Improve the Prediction of Osteoporotic Fractures. Endocrinol Metab (Seoul) 2020; 35:55-63. [PMID: 32207264 PMCID: PMC7090300 DOI: 10.3803/enm.2020.35.1.55] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/03/2019] [Accepted: 12/31/2019] [Indexed: 12/27/2022] Open
Abstract
Osteoporotic fracture (OF) is associated with high disability and morbidity rates. The burden of OF may be reduced by early identification of subjects who are vulnerable to fracture. Although the current fracture risk assessment model includes clinical risk factors (CRFs) and bone mineral density (BMD), its overall ability to identify individuals at high risk for fracture remains suboptimal. Efforts have therefore been made to identify potential biomarkers that can predict the risk of OF, independent of or combined with CRFs and BMD. This review highlights the emerging biomarkers of bone metabolism, including sphongosine-1-phosphate, leucine-rich repeat-containing 17, macrophage migration inhibitory factor, sclerostin, receptor activator of nuclear factor-κB ligand, and periostin, and the importance of biomarker risk score, generated by combining these markers, in enhancing the accuracy of fracture prediction.
Collapse
Affiliation(s)
- Beom Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
283
|
Leitch VD, Bassett JHD, Williams GR. Role of thyroid hormones in craniofacial development. Nat Rev Endocrinol 2020; 16:147-164. [PMID: 31974498 DOI: 10.1038/s41574-019-0304-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
Abstract
The development of the craniofacial skeleton relies on complex temporospatial organization of diverse cell types by key signalling molecules. Even minor disruptions to these processes can result in deleterious consequences for the structure and function of the skull. Thyroid hormone deficiency causes delayed craniofacial and tooth development, dysplastic facial features and delayed development of the ossicles in the middle ear. Thyroid hormone excess, by contrast, accelerates development of the skull and, in severe cases, might lead to craniosynostosis with neurological sequelae and facial hypoplasia. The pathogenesis of these important abnormalities remains poorly understood and underinvestigated. The orchestration of craniofacial development and regulation of suture and synchondrosis growth is dependent on several critical signalling pathways. The underlying mechanisms by which these key pathways regulate craniofacial growth and maturation are largely unclear, but studies of single-gene disorders resulting in craniofacial malformations have identified a number of critical signalling molecules and receptors. The craniofacial consequences resulting from gain-of-function and loss-of-function mutations affecting insulin-like growth factor 1, fibroblast growth factor receptor and WNT signalling are similar to the effects of altered thyroid status and mutations affecting thyroid hormone action, suggesting that these critical pathways interact in the regulation of craniofacial development.
Collapse
Affiliation(s)
- Victoria D Leitch
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Royal Melbourne Institute of Technology (RMIT) Centre for Additive Manufacturing, RMIT University, Melbourne, VIC, Australia
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
284
|
Komori T. Molecular Mechanism of Runx2-Dependent Bone Development. Mol Cells 2020; 43:168-175. [PMID: 31896233 PMCID: PMC7057844 DOI: 10.14348/molcells.2019.0244] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 12/03/2019] [Indexed: 01/09/2023] Open
Abstract
Runx2 is an essential transcription factor for skeletal development. It is expressed in multipotent mesenchymal cells, osteoblast-lineage cells, and chondrocytes. Runx2 plays a major role in chondrocyte maturation, and Runx3 is partly involved. Runx2 regulates chondrocyte proliferation by directly regulating Ihh expression. It also determines whether chondrocytes become those that form transient cartilage or permanent cartilage, and functions in the pathogenesis of osteoarthritis. Runx2 is essential for osteoblast differentiation and is required for the proliferation of osteoprogenitors. Ihh is required for Runx2 expression in osteoprogenitors, and hedgehog signaling and Runx2 induce the differentiation of osteoprogenitors to preosteoblasts in endochondral bone. Runx2 induces Sp7 expression, and Runx2, Sp7, and canonical Wnt signaling are required for the differentiation of preosteoblasts to immature osteoblasts. It also induces the proliferation of osteoprogenitors by directly regulating the expression of Fgfr2 and Fgfr3. Furthermore, Runx2 induces the proliferation of mesenchymal cells and their commitment into osteoblast-lineage cells through the induction of hedgehog (Gli1, Ptch1, Ihh), Fgf (Fgfr2, Fgfr3), Wnt (Tcf7, Wnt10b), and Pthlh (Pth1r) signaling pathway gene expression in calvaria, and more than a half-dosage of Runx2 is required for their expression. This is a major cause of cleidocranial dysplasia, which is caused by heterozygous mutation of RUNX2. Cbfb, which is a co-transcription factor that forms a heterodimer with Runx2, enhances DNA binding of Runx2 and stabilizes Runx2 protein by inhibiting its ubiquitination. Thus, Runx2/Cbfb regulates the proliferation and differentiation of chondrocytes and osteoblast-lineage cells by activating multiple signaling pathways and via their reciprocal regulation.
Collapse
Affiliation(s)
- Toshihisa Komori
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| |
Collapse
|
285
|
Wang C, Qiao X, Zhang Z, Li C. MiR-128 promotes osteogenic differentiation of bone marrow mesenchymal stem cells in rat by targeting DKK2. Biosci Rep 2020; 40:BSR20182121. [PMID: 31985779 PMCID: PMC7007406 DOI: 10.1042/bsr20182121] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/07/2020] [Accepted: 01/15/2020] [Indexed: 02/05/2023] Open
Abstract
Bone loss caused by inflammatory disease, such as peri-implantitis, poses a great challenge to clinicians for restoration. Emerging evidence indicates that microRNAs (miRNAs) are indispensable regulators of bone growth, development, and formation. In the present study, we found that microRNA-128 (miR-128) was differentially up-regulated during the osteogenic differentiation of rat bone marrow stem cells (rBMSCs). Overexpression of miR-128 promoted osteogenic differentiation of rBMSCs by up-regulating alkaline phosphatase (ALP), matrix mineralization, mRNA, and protein levels of osteogenic makers (e.g. RUNX2, BMP-2, and COLIA1), whereas inhibition of miR-128 suppressed osteoblastic differentiation in vitro. Mechanistically, miR-128 directly and functionally targeted Dickkopf2 (DKK2), which is a Wnt signaling pathway antagonist, and enhanced Wnt/β-catenin signaling activity. Furthermore, the positive effect of miR-128 on osteogenic differentiation was apparently abrogated by DKK2 overexpression. Collectively, these results indicate that miR-128 promotes osteogenic differentiation of rBMSCs by targeting DKK2, which may provide a promising approach to the treatment of peri-implantitis.
Collapse
Affiliation(s)
- Can Wang
- Department of Head and Neck Oncology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xianghe Qiao
- Department of Head and Neck Oncology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhuang Zhang
- Department of Head and Neck Oncology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chunjie Li
- Department of Head and Neck Oncology, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
286
|
Guo D, He H, Zhao M, Zhang G, Hou T. Desalted duck egg white peptides promoted osteogenesis via wnt/β-catenin signal pathway. J Food Sci 2020; 85:834-842. [PMID: 32078745 DOI: 10.1111/1750-3841.15067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/07/2020] [Accepted: 01/17/2020] [Indexed: 12/18/2022]
Abstract
Osteoporosis is a degenerative disease that threatens bone health of the elderly (especially postmenopausal women). Since osteoporosis is important to prevent, the aim of this study was to investigate the regulation of desalted duck egg white peptides (DPs) on osteoporosis. In this study, the effects of DPs on bone formation were evaluated using MC3T3-E1 cells and ovariectomized (OVX) rats. DPs significantly enhanced the preosteoblasts proliferation, differentiation, and matrix mineralization via the upregulation of wnt3a expression, low-density lipoprotein receptor-related protein-5 (LRP-5), β-catenin, runt-related transcription factor 2 (Runx2), and osteoprotegerin (OPG) (P < 0.05). The intracellular calcium concentration was significantly elevated by DPs (P < 0.05), which is attributed to calcium influx and L-type calcium channels. Additionally, OVX rat model experiment indicated that DPs (600 mg/kg bw) had a superior effect against bone loss induced by estrogen deficiency, as it significantly declined bone turnover markers, and significantly increased biomechanical parameters (P < 0.05). Mineralized bone surfaces and bone microstructure were also obviously improved by DPs treatment. Immunohistochemical analysis showed that receptor activator of nuclear factor κ B (RANK) expression of tibia in DPs group was significantly reduced compared with the model group (P < 0.05). Our results demonstrated that DPs could enhance preosteoblasts differentiation and antiosteoporosis via wnt/β-catenin signal pathway and several key osteogenic transcription factors such as Runx2 and OPG. PRACTICAL APPLICATION: High-value utilization of salted duck egg white, a byproduct of food industry, is worthy of in-depth study. Desalted duck egg white peptides (DPs) were proved to promote bone formation, which suggests the potentials of DPs as cofactors in osteoporosis prevention.
Collapse
Affiliation(s)
- Danjun Guo
- College of Food Science and Technology, Huazhong Agricultural Univ., Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural Univ.), Ministry of Education, Wuhan, 43000, China
| | - Hui He
- College of Food Science and Technology, Huazhong Agricultural Univ., Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural Univ.), Ministry of Education, Wuhan, 43000, China
| | - Mengge Zhao
- College of Food Science and Technology, Huazhong Agricultural Univ., Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural Univ.), Ministry of Education, Wuhan, 43000, China
| | - Guoqing Zhang
- College of Food Science and Technology, Huazhong Agricultural Univ., Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural Univ.), Ministry of Education, Wuhan, 43000, China
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural Univ., Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural Univ.), Ministry of Education, Wuhan, 43000, China
| |
Collapse
|
287
|
Arroyo R, López S, Romo E, Montoya G, Hoz L, Pedraza C, Garfias Y, Arzate H. Carboxy-Terminal Cementum Protein 1-Derived Peptide 4 (cemp1-p4) Promotes Mineralization through wnt/ β-catenin Signaling in Human Oral Mucosa Stem Cells. Int J Mol Sci 2020; 21:E1307. [PMID: 32075221 PMCID: PMC7072908 DOI: 10.3390/ijms21041307] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022] Open
Abstract
Human cementum protein 1 (CEMP1) is known to induce cementoblast and osteoblast differentiation and alkaline phosphatase (ALP) activity in human periodontal ligament-derived cells in vitro and promotes bone regeneration in vivo. CEMP1's secondary structure analysis shows that it has a random-coiled structure and is considered an Intrinsic Disordered Protein (IDP). CEMP1's short peptide sequences mimic the biological capabilities of CEMP1. However, the role and mechanisms of CEMP1's C-terminal-derived synthetic peptide (CEMP1-p4) in the canonical Wnt/β-catenin signaling pathway are yet to be described. Here we report that CEMP1-p4 promotes proliferation and differentiation of Human Oral Mucosa Stem Cells (HOMSCs) by activating the Wnt/β-catenin pathway. CEMP1-p4 stimulation upregulated the expression of β-catenin and glycogen synthase kinase 3 beta (GSK-3B) and activated the transcription factors TCF1/7 and Lymphoid Enhancer binding Factor 1 (LEF1) at the mRNA and protein levels. We found translocation of β-catenin to the nucleus in CEMP1-p4-treated cultures. The peptide also penetrates the cell membrane and aggregates around the cell nucleus. Analysis of CEMP1-p4 secondary structure revealed that it has a random-coiled structure. Its biological activities included the induction to nucleate hydroxyapatite crystals. In CEMP1-p4-treated HOMSCs, ALP activity and calcium deposits increased. Expression of Osterix (OSX), Runt-related transcription factor 2 (RUNX2), Integrin binding sialoproptein (IBSP) and osteocalcin (OCN) were upregulated. Altogether, these data show that CEMP1-p4 plays a direct role in the differentiation of HOMSCs to a "mineralizing-like" phenotype by activating the β-catenin signaling cascade.
Collapse
Affiliation(s)
- Rita Arroyo
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico; (R.A.); (S.L.); (E.R.); (G.M.); (L.H.); (C.P.)
| | - Sonia López
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico; (R.A.); (S.L.); (E.R.); (G.M.); (L.H.); (C.P.)
| | - Enrique Romo
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico; (R.A.); (S.L.); (E.R.); (G.M.); (L.H.); (C.P.)
| | - Gonzalo Montoya
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico; (R.A.); (S.L.); (E.R.); (G.M.); (L.H.); (C.P.)
| | - Lía Hoz
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico; (R.A.); (S.L.); (E.R.); (G.M.); (L.H.); (C.P.)
| | - Claudia Pedraza
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico; (R.A.); (S.L.); (E.R.); (G.M.); (L.H.); (C.P.)
| | - Yonathan Garfias
- Departamento de Bioquímica, Facultad de Medicina, UNAM, Universidad Nacional Autónoma de México, CDMX 04510, Mexico;
- Instituto de Oftalmología Conde de Valenciana, CDMX 06800, Mexico
| | - Higinio Arzate
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad Nacional Autónoma de México, CDMX 04510, Mexico; (R.A.); (S.L.); (E.R.); (G.M.); (L.H.); (C.P.)
| |
Collapse
|
288
|
Abstract
Post-natal bone development is characterized by substantial longitudinal bone growth and changes in skeletal size and shape. Bone is in a dynamic process of continuous remodeling which helps to regulate calcium homeostasis, repair micro-damage to bones from everyday stress, and to shape the skeleton during growth. Bone growth is regulated by systemic hormones and locally generated factors. Understanding their mechanisms of action enables us to obtain a better appreciation of the cellular and molecular basis of bone remodeling and could therefore be valuable in approaches to new therapies. This article will review molecular and cellular control of skeletal growth in the post-natal period, the physiology of each bone cell with their systemic and local regulators, as well as the physiology of bone remodeling.
Collapse
Affiliation(s)
- Rania Ali El-Farrash
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| | - Radwa Hassan Ali
- Physiology Department, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| | - Noha Mokhtar Barakat
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Abbassya Square, 11566, Cairo, Egypt.
| |
Collapse
|
289
|
Kim K, Kim JH, Kim I, Seong S, Kim N. Rev-erbα Negatively Regulates Osteoclast and Osteoblast Differentiation through p38 MAPK Signaling Pathway. Mol Cells 2020; 43:34-47. [PMID: 31896234 PMCID: PMC6999712 DOI: 10.14348/molcells.2019.0232] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/21/2019] [Accepted: 12/01/2019] [Indexed: 11/27/2022] Open
Abstract
The circadian clock regulates various physiological processes, including bone metabolism. The nuclear receptors Reverbs, comprising Rev-erbα and Rev-erbβ, play a key role as transcriptional regulators of the circadian clock. In this study, we demonstrate that Rev-erbs negatively regulate differentiation of osteoclasts and osteoblasts. The knockdown of Rev-erbα in osteoclast precursor cells enhanced receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation, as well as expression of nuclear factor of activated T cells 1 (NFATc1), osteoclast-associated receptor (OSCAR), and tartrate-resistant acid phosphatase (TRAP). The overexpression of Rev-erbα leads to attenuation of the NFATc1 expression via inhibition of recruitment of c-Fos to the NFATc1 promoter. The overexpression of Rev-erbα in osteoblast precursors attenuated the expression of osteoblast marker genes including Runx2, alkaline phosphatase (ALP), bone sialoprotein (BSP), and osteocalcin (OC). Rev-erbα interfered with the recruitment of Runx2 to the promoter region of the target genes. Conversely, knockdown of Reverbα in the osteoblast precursors enhanced the osteoblast differentiation and function. In addition, Rev-erbα negatively regulated osteoclast and osteoblast differentiation by suppressing the p38 MAPK pathway. Furthermore, intraperitoneal administration of GSK4112, a Rev-erb agonist, protects RANKL-induced bone loss via inhibition of osteoclast differentiation in vivo . Taken together, our results demonstrate a molecular mechanism of Rev-erbs in the bone remodeling, and provide a molecular basis for a potential therapeutic target for treatment of bone disease characterized by excessive bone resorption.
Collapse
MESH Headings
- Animals
- Bone Remodeling
- Bone Resorption/genetics
- Bone Resorption/metabolism
- Cell Differentiation
- Cells, Cultured
- Circadian Clocks
- Disease Models, Animal
- Gain of Function Mutation/genetics
- Humans
- Male
- Mice
- Mice, Inbred ICR
- Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Osteoblasts/physiology
- Osteoclasts/physiology
- Osteogenesis/genetics
- RNA, Small Interfering/genetics
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469,
Korea
| |
Collapse
|
290
|
Identification of RELN variant p.(Ser2486Gly) in an Iranian family with ankylosing spondylitis; the first association of RELN and AS. Eur J Hum Genet 2020; 28:754-762. [PMID: 32001840 DOI: 10.1038/s41431-020-0573-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/11/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Ankylosing spondylitis (AS) is a common complex inflammatory disease; however, up to now distinct genes with monogenic pattern have not been reported for this disease. In the present study, we report a large Iranian family with several affected members with AS. DNAs of the three affected and two healthy cases were chosen for performing whole-exome sequencing (WES). After several filtering steps, candidate variants in the following genes were detected: RELN, DNMT1, TAF4β, MUC16, DLG2, and FAM208. However, segregation analysis confirmed the association of only one variant, c.7456A>G; p.(Ser2486Gly) in the RELN gene with AS in this family. In addition, in silico predictions supported the probable pathogenicity of this variant. In this study, for the first time, we report a novel variant in the RELN gene, c.7456A>G; p.(Ser2486Gly), which completely co-segregates with AS. This association suggests potential insights into the pathophysiological bases of AS and it could broaden horizons toward new therapeutic strategies.
Collapse
|
291
|
Russo C, Ferro Y, Maurotti S, Salvati MA, Mazza E, Pujia R, Terracciano R, Maggisano G, Mare R, Giannini S, Romeo S, Pujia A, Montalcini T. Lycopene and bone: an in vitro investigation and a pilot prospective clinical study. J Transl Med 2020; 18:43. [PMID: 31996227 PMCID: PMC6990577 DOI: 10.1186/s12967-020-02238-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/22/2020] [Indexed: 12/12/2022] Open
Abstract
Background There are several effective therapies for osteoporosis but these agents might cause serious adverse events. Lycopene intake could prevent bone loss, however studies on its effects on bone are scarce. Our aim was to investigate the effects of lycopene on osteoblast cells as well as bone mineral density and bone turnover markers in postmenopausal women. Methods We investigated the effect of lycopene on the Wnt/β-catenin and ERK 1/2 pathways, RUNX2, alkaline phosphatase, RANKL and COL1A of Saos-2. We also carried out a pilot controlled clinical study to verify the feasibility of an approach for bone loss prevention through the intake of a lycopene-rich tomato sauce in 39 postmenopausal women. Results Lycopene 10 µM resulted in higher β-catenin and phERK1/2 protein Vs the vehicle (p = 0.04 and p = 0.006). RUNX2 and COL1A mRNA was induced by both 5 and 10 µM doses (p = 0.03; p = 0.03 and p = 0.03; p = 0.05) while RANKL mRNA was reduced (p < 0.05). A significant bone density loss was not detected in women taking the tomato sauce while the control group had bone loss (p = 0.002). Tomato sauce intake resulted in a greater bone alkaline phosphatase reduction than the control (18% vs 8.5%, p = 0.03). Conclusions Lycopene activates the WNT/β-catenin and ERK1/2 pathways, upregulates RUNX2, alkaline phosphatase, COL1A and downregulates RANKL Saos-2. These processes contributed to prevent bone loss in postmenopausal women.
Collapse
Affiliation(s)
- Cristina Russo
- Department of Clinical and Experimental Medicine, Nutrition Unit, University Magna Grecia, 88100, Catanzaro, Italy
| | - Yvelise Ferro
- Department of Health Science, University Magna Graecia, 88100, Catanzaro, Italy
| | - Samantha Maurotti
- Department of Medical and Surgical Science, University Magna Graecia, 88100, Catanzaro, Italy
| | | | - Elisa Mazza
- Department of Medical and Surgical Science, University Magna Graecia, 88100, Catanzaro, Italy
| | - Roberta Pujia
- Department of Medical and Surgical Science, University Magna Graecia, 88100, Catanzaro, Italy
| | - Rosa Terracciano
- Department of Health Science, University Magna Graecia, 88100, Catanzaro, Italy
| | | | - Rosario Mare
- Department of Clinical and Experimental Medicine, Nutrition Unit, University Magna Grecia, 88100, Catanzaro, Italy
| | - Sandro Giannini
- Department of Medicine, Clinica Medica 1, University of Padova and Regional Centre for Osteoporosis, Padua, Italy
| | - Stefano Romeo
- Department of Medical and Surgical Science, University Magna Graecia, 88100, Catanzaro, Italy.,Department of Molecular and Clinical Medicine, Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, 42246, Göteborg, Sweden
| | - Arturo Pujia
- Department of Medical and Surgical Science, University Magna Graecia, 88100, Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Clinical and Experimental Medicine, Nutrition Unit, University Magna Grecia, 88100, Catanzaro, Italy.
| |
Collapse
|
292
|
A Wnt-mediated transformation of the bone marrow stromal cell identity orchestrates skeletal regeneration. Nat Commun 2020; 11:332. [PMID: 31949165 PMCID: PMC6965122 DOI: 10.1038/s41467-019-14029-w] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022] Open
Abstract
Bone marrow stromal cells (BMSCs) are versatile mesenchymal cell populations underpinning the major functions of the skeleton, a majority of which adjoin sinusoidal blood vessels and express C-X-C motif chemokine ligand 12 (CXCL12). However, how these cells are activated during regeneration and facilitate osteogenesis remains largely unknown. Cell-lineage analysis using Cxcl12-creER mice reveals that quiescent Cxcl12-creER+ perisinusoidal BMSCs differentiate into cortical bone osteoblasts solely during regeneration. A combined single cell RNA-seq analysis demonstrate that these cells convert their identity into a skeletal stem cell-like state in response to injury, associated with upregulation of osteoblast-signature genes and activation of canonical Wnt signaling components along the single-cell trajectory. β-catenin deficiency in these cells indeed causes insufficiency in cortical bone regeneration. Therefore, quiescent Cxcl12-creER+ BMSCs transform into osteoblast precursor cells in a manner mediated by canonical Wnt signaling, highlighting a unique mechanism by which dormant stromal cells are enlisted for skeletal regeneration. Bone marrow stromal cells (BMSCs) lining sinusoidal blood vessels are mesenchymal cells whose function is critical for the skeleton. Here the authors show that quiescent CXCL12-expressing BMSCs can convert into a skeletal stem cell-like state, and differentiate into cortical bone osteoblasts only in response to injury.
Collapse
|
293
|
Zujur D, Kanke K, Onodera S, Tani S, Lai J, Azuma T, Xin X, Lichtler AC, Rowe DW, Saito T, Tanaka S, Masaki H, Nakauchi H, Chung UI, Hojo H, Ohba S. Stepwise strategy for generating osteoblasts from human pluripotent stem cells under fully defined xeno-free conditions with small-molecule inducers. Regen Ther 2020; 14:19-31. [PMID: 31988991 PMCID: PMC6965656 DOI: 10.1016/j.reth.2019.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/20/2019] [Accepted: 12/24/2019] [Indexed: 01/01/2023] Open
Abstract
Clinically relevant human induced pluripotent stem cell (hiPSC) derivatives require efficient protocols to differentiate hiPSCs into specific lineages. Here we developed a fully defined xeno-free strategy to direct hiPSCs toward osteoblasts within 21 days. The strategy successfully achieved the osteogenic induction of four independently derived hiPSC lines by a sequential use of combinations of small-molecule inducers. The induction first generated mesodermal cells, which subsequently recapitulated the developmental expression pattern of major osteoblast genes and proteins. Importantly, Col2.3-Cherry hiPSCs subjected to this strategy strongly expressed the cherry fluorescence that has been observed in bone-forming osteoblasts in vivo. Moreover, the protocol combined with a three-dimensional (3D) scaffold was suitable for the generation of a xeno-free 3D osteogenic system. Thus, our strategy offers a platform with significant advantages for bone biology studies and it will also contribute to clinical applications of hiPSCs to skeletal regenerative medicine.
Collapse
Affiliation(s)
- Denise Zujur
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Kosuke Kanke
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, Tokyo, Japan
| | - Shoichiro Tani
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jenny Lai
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Toshifumi Azuma
- Department of Biochemistry, Tokyo Dental College, Tokyo, Japan
| | - Xiaonan Xin
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Alexander C Lichtler
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - David W Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Taku Saito
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideki Masaki
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ung-Il Chung
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hironori Hojo
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinsuke Ohba
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
294
|
Disruption of Dhcr7 and Insig1/2 in cholesterol metabolism causes defects in bone formation and homeostasis through primary cilium formation. Bone Res 2020; 8:1. [PMID: 31934493 PMCID: PMC6946666 DOI: 10.1038/s41413-019-0078-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023] Open
Abstract
Human linkage studies suggest that craniofacial deformities result from either genetic mutations related to cholesterol metabolism or high-cholesterol maternal diets. However, little is known about the precise roles of intracellular cholesterol metabolism in the development of craniofacial bones, the majority of which are formed through intramembranous ossification. Here, we show that an altered cholesterol metabolic status results in abnormal osteogenesis through dysregulation of primary cilium formation during bone formation. We found that cholesterol metabolic aberrations, induced through disruption of either Dhcr7 (which encodes an enzyme involved in cholesterol synthesis) or Insig1 and Insig2 (which provide a negative feedback mechanism for cholesterol biosynthesis), result in osteoblast differentiation abnormalities. Notably, the primary cilia responsible for sensing extracellular cues were altered in number and length through dysregulated ciliary vesicle fusion in Dhcr7 and Insig1/2 mutant osteoblasts. As a consequence, WNT/β-catenin and hedgehog signaling activities were altered through dysregulated primary cilium formation. Strikingly, the normalization of defective cholesterol metabolism by simvastatin, a drug used in the treatment of cholesterol metabolic aberrations, rescued the abnormalities in both ciliogenesis and osteogenesis in vitro and in vivo. Thus, our results indicate that proper intracellular cholesterol status is crucial for primary cilium formation during skull formation and homeostasis.
Collapse
|
295
|
Ibarra BA, Atit R. What Do Animal Models Teach Us About Congenital Craniofacial Defects? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:137-155. [PMID: 32304072 PMCID: PMC7394376 DOI: 10.1007/978-981-15-2389-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The formation of the head and face is a complex process which involves many different signaling cues regulating the migration, differentiation, and proliferation of the neural crest. This highly complex process is very error-prone, resulting in craniofacial defects in nearly 10,000 births in the United States annually. Due to the highly conserved mechanisms of craniofacial development, animal models are widely used to understand the pathogenesis of various human diseases and assist in the diagnosis and generation of preventative therapies and treatments. Here, we provide a brief background of craniofacial development and discuss several rare diseases affecting craniofacial bone development. We focus on rare congenital diseases of the cranial bone, facial jaw bones, and two classes of diseases, ciliopathies and RASopathies. Studying the animal models of these rare diseases sheds light not only on the etiology and pathology of each disease, but also provides meaningful insights towards the mechanisms which regulate normal development of the head and face.
Collapse
Affiliation(s)
- Beatriz A Ibarra
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Radhika Atit
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA.
- Department of Genetics, Case Western Reserve University, Cleveland, OH, USA.
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
296
|
Lv Y, Huang Y, Xu M, Heng BC, Yang C, Cao C, Hu Z, Liu W, Chi X, Gao M, Zhang X, Wei Y, Deng X. The miR-193a-3p-MAP3k3 Signaling Axis Regulates Substrate Topography-Induced Osteogenesis of Bone Marrow Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901412. [PMID: 31921551 PMCID: PMC6947707 DOI: 10.1002/advs.201901412] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 09/30/2019] [Indexed: 06/10/2023]
Abstract
Substrate topographical features induce osteogenic differentiation of bone marrow stem cells (BMSCs), but the underlying mechanisms are unclear. As microRNAs (miRNAs) play key roles in osteogenesis and bone regeneration, it would be meaningful to elucidate the roles of miRNAs in the intracellular signaling cascade of topographical cue-induced osteogenic differentiation. In this study, the miRNA expression profile of the topographical feature-induced osteogenic differentiation group is different from that of the chemical-factors-induced osteogenic differentiation group. miR-193a-3p is sensitive to substrate topographical features and its downregulation enhances osteogenic differentiation only in the absence of osteogenesis-inducing medium. Also, substrate topographical features specifically activate a nonclassical osteogenetic pathway-the mitogen-activated protein kinase (MAPK) pathway. Loss- and gain-of-function experiments demonstrate that miR-193a-3p regulates the MAPK pathway by targeting the MAP3k3 gene. In conclusion, this data indicates that different osteogenic-lineage-related intracellular signaling cascades are triggered in BMSCs subjected to biophysical or chemical stimulation. Moreover, the miR-193a-3p-MAP3k3 signaling axis plays a pivotal role in the transduction of biophysical cues from the substrate to regulate the osteogenic lineage specification of BMSCs, and hence may be a promising molecular target for bone regenerative therapies.
Collapse
Affiliation(s)
- Yan Lv
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Ying Huang
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Mingming Xu
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Boon Chin Heng
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Congchong Yang
- Department of Cariology and EndodontologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Cen Cao
- Department of StomatologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Zhewen Hu
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Wenwen Liu
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiaopei Chi
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Min Gao
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuehui Zhang
- Department of Dental Materials and Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yan Wei
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Department of Geriatric DentistryNMPA Key Laboratory for Dental MaterialsNational Engineering Laboratory for Digital and Material Technology of StomatologyBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
297
|
Hsiao CY, Chen TH, Chu TH, Ting YN, Tsai PJ, Shyu JF. Calcitonin Induces Bone Formation by Increasing Expression of Wnt10b in Osteoclasts in Ovariectomy-Induced Osteoporotic Rats. Front Endocrinol (Lausanne) 2020; 11:613. [PMID: 33013696 PMCID: PMC7506163 DOI: 10.3389/fendo.2020.00613] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/27/2020] [Indexed: 11/13/2022] Open
Abstract
Calcitonin is a small peptide hormone secreted from the parafollicular cells of the thyroid gland in response to an increase in serum calcium. The inhibition of osteoclastic resorption is the main mechanism by which calcitonin quickly decreases circulating calcium levels. Although calcitonin pharmacologically acts on osteoclasts to prevent bone resorption, the results of studies on genetically modified animals have shown that the physiological effect of calcitonin is in the inhibition of osteoblastic bone formation. Because the calcitonin receptor is only expressed in osteoclasts, the effect of calcitonin on osteoblasts maybe indirect and mediated via osteoclasts. Wnt ligands are involved in various aspects of skeletal biology, including bone remodeling and endochondral bone formation. Wnt10b has recently been recognized as a clastokine, and is potentially a therapeutic target for treating bone disorders. However, the extent to which Wnt signaling is involved in bone physiology and disease is not yet fully understood. We hypothesize that calcitonin indirectly increases osteoblastic bone formation by inducing Wnt10b expression in osteoclasts. Micro-CT analysis revealed reduced bone loss in calcitonin-treated ovariectomized rats. The serum of animals treated with calcitonin had decreased TRAP5b and CTX-1 but increased osteocalcin, P1NP, and Wnt10b. Immunohistochemistry staining showed that the level of Wnt10b in the femur was increased in calcitonin-treated groups as compared with control groups. Hematopoietic mononuclear cells were separated from rat femur and tibia bone marrow, and were induced into osteoclasts following treatment with M-CSF and RANKL. In these cells, immunoconfocal microscopy and Western blot analysis showed that calcitonin induced an increase in Wnt10b expression. In a culture of osteoblasts isolated from neonatal rat calvariae, the calcitonin-treated osteoclast supernatant showed an increase in mineralization, as indicated by ALP and alizarin red staining. Taken together, these results indicate that calcitonin induces bone formation by increasing the expression of Wnt10b in osteoclasts in ovariectomy-induced osteoporotic rats. The present study provides in-depth information about the effects of calcitonin on bone remodeling and will thus help in the development of future potential therapeutic strategies for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Chen-Yuan Hsiao
- National Defense Medical Center, Graduate Institute of Medical Sciences, Taipei, Taiwan
- Department of Surgery, Landseed International Hospital, Taoyuan, Taiwan
| | - Tien-Hua Chen
- School of Medicine, Institute of Anatomy and Cell Biology, National Yang Ming University, Taipei, Taiwan
- Department of Surgery, Trauma Center, Veterans General Hospital, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Hui Chu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yen-Nien Ting
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Pei-Jiun Tsai
- School of Medicine, Institute of Anatomy and Cell Biology, National Yang Ming University, Taipei, Taiwan
- Department of Surgery, Trauma Center, Veterans General Hospital, Taipei, Taiwan
- Department of Critical Care Medicine, Veterans General Hospital, Taipei, Taiwan
- *Correspondence: Pei-Jiun Tsai
| | - Jia-Fwu Shyu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
- Department of Psychiatry, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
- Jia-Fwu Shyu
| |
Collapse
|
298
|
Silva MJ, Holguin N. Aging aggravates intervertebral disc degeneration by regulating transcription factors toward chondrogenesis. FASEB J 2019; 34:1970-1982. [PMID: 31909538 DOI: 10.1096/fj.201902109r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/08/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022]
Abstract
Osterix is a critical transcription factor of mesenchymal stem cell fate, where its loss or loss of Wnt signaling diverts differentiation to a chondrocytic lineage. Intervertebral disc (IVD) degeneration activates the differentiation of prehypertrophic chondrocyte-like cells and inactivates Wnt signaling, but its interactive role with osterix is unclear. First, compared to young-adult (5 mo), mechanical compression of old (18 mo) IVD induced greater IVD degeneration. Aging (5 vs 12 mo) and/or compression reduced the transcription of osterix and notochordal marker T by 40-75%. Compression elevated the transcription of hypertrophic chondrocyte marker MMP13 and pre-osterix transcription factor RUNX2, but less so in 12 mo IVD. Next, using an Ai9/td reporter and immunohistochemical staining, annulus fibrosus and nucleus pulposus cells of young-adult IVD expressed osterix, but aging and compression reduced its expression. Lastly, in vivo LRP5-deficiency in osterix-expressing cells inactivated Wnt signaling in the nucleus pulposus by 95%, degenerated the IVD to levels similar to aging and compression, reduced the biomechanical properties by 45-70%, and reduced the transcription of osterix, notochordal markers and chondrocytic markers by 60-80%. Overall, these data indicate that age-related inactivation of Wnt signaling in osterix-expressing cells may limit regeneration by depleting the progenitors and attenuating the expansion of chondrocyte-like cells.
Collapse
Affiliation(s)
- Matthew J Silva
- Department of Biomedical Engineering, Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, USA
| | - Nilsson Holguin
- Department of Mechanical and Energy Engineering, Indiana Center for Musculoskeletal Health, IUPUI, Indianapolis, IN, USA.,Department of Anatomy and Cell Biology, Indiana Center for Musculoskeletal Health, IUPUI, Indianapolis, IN, USA
| |
Collapse
|
299
|
Chen D, Kim DJ, Shen J, Zou Z, O'Keefe RJ. Runx2 plays a central role in Osteoarthritis development. J Orthop Translat 2019; 23:132-139. [PMID: 32913706 PMCID: PMC7452174 DOI: 10.1016/j.jot.2019.11.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis, is the leading cause of impaired mobility in the elderly, and accounts for more than a third of chronic moderate to severe pain. As a degenerative joint disorder, OA affects the whole joint and results in synovial hyperplasia, degradation of articular cartilage, subchondral sclerosis, osteophyte formation, and chronic pain. Currently, there is no effective drug to decelerate OA progression and molecular targets for drug development have been insufficiently investigated. Anti-OA drug development can benefit from more and precise knowledge of molecular targets for drug development. Runt-related transcription factor 2 (Runx2) is a key transcription factor controlling osteoblast and chondrocyte differentiation and is among the most promising potential therapeutic targets. Notably, Runx2 expression is upregulated in several murine OA models, suggesting a role in disease pathogenesis. In this review article, we summarized recent findings on Runx2 related to OA development and evaluated its potential as a therapeutic target. The translational potential of this article A better understanding of the role of Runx2 in osteoarthritis pathogenesis will contribute to the development of novel intervention of osteoarthritis disease.
Collapse
Affiliation(s)
- Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dongyeon J Kim
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| | - Jie Shen
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| | - Zhen Zou
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| | - Regis J O'Keefe
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| |
Collapse
|
300
|
Huang J, Cai X, Ou Y, Fan L, Zhou Y, Wang Y. Protective roles of FICZ and aryl hydrocarbon receptor axis on alveolar bone loss and inflammation in experimental periodontitis. J Clin Periodontol 2019; 46:882-893. [PMID: 31286538 DOI: 10.1111/jcpe.13166] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 03/21/2019] [Accepted: 07/02/2019] [Indexed: 12/27/2022]
Abstract
AIM The aryl hydrocarbon receptor (AhR)-ligand axis has been shown to be involved in inflammatory diseases and bone homeostasis. However, the activation of AhR signalling pathway and the possible functions of AhR ligands in periodontitis are underexplored. This study investigated the expression of the AhR target gene cytochrome P450 subfamily B member 1 (CYP1B1) and the functions and mechanisms of the AhR ligand 6 formylindolo[3,2-b]carbazole (FICZ) in periodontitis. MATERIALS AND METHODS CYP1B1 expression was detected in human periodontitis samples, mice with ligature-induced periodontitis and lipopolysaccharide (LPS)-induced inflammation in periodontal ligament cells (PDLCs) in vitro. FICZ was administered topically or systemically. The therapeutic functions of FICZ were detected via qPCR, micro-computed tomography and immunohistochemistry. Finally, the mechanisms of AhR signalling in periodontitis were investigated by cell assays. RESULTS CYP1B1 expression was downregulated in periodontitis. FICZ rescued the alveolar bone loss and mitigated the inflammatory cytokines in periodontitis mice. In vitro, FICZ pre-treatment reduced the LPS-induced inflammation in PDLCs via the increased phosphorylation of STAT3. Additionally, FICZ prompted the mineralization of PDLCs via activation of the Wnt/β-catenin signalling pathway. CONCLUSION AhR signalling pathway is suppressed in periodontitis and the AhR ligand FICZ can prevent periodontitis.
Collapse
Affiliation(s)
- Jing Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xinjie Cai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Prosthodontics, Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yanjing Ou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Le Fan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Prosthodontics, Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yining Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Prosthodontics, Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|