1
|
Uptegrove A, Chen C, Sahagun-Bisson M, Kulkarni AK, Louie KW, Ueharu H, Mishina Y, Omi-Sugihara M. Influence of bone morphogenetic protein (BMP) signaling and masticatory load on morphological alterations of the mouse mandible during postnatal development. Arch Oral Biol 2025; 169:106096. [PMID: 39341045 PMCID: PMC11609011 DOI: 10.1016/j.archoralbio.2024.106096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/15/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE Bone homeostasis relies on several contributing factors, encompassing growth factors and mechanical stimuli. While bone morphogenetic protein (BMP) signaling is acknowledged for its essential role in skeletal development, its specific impact on mandibular morphogenesis remains unexplored. Here, we investigated the involvement of BMP signaling and mechanical loading through mastication in postnatal mandibular morphogenesis. DESIGN We employed conditional deletion of Bmpr1a in osteoblasts and chondrocytes via Osterix-Cre. Cre activity was induced at birth for the 3-week group and at three weeks for the 9-week and 12-week groups, respectively. The conditional knockout (cKO) and control mice were given either a regular diet (hard diet, HD) or a powdered diet (soft diet, SD) from 3 weeks until sample collection, followed by micro-CT and histological analysis. RESULTS The cKO mice exhibited shorter anterior lengths and a posteriorly inclined ramus across all age groups compared to the control mice. The cKO mice displayed an enlarged hypertrophic cartilage area along with fewer osteoclast numbers in the subchondral bone of the condyle compared to the control group at three weeks, followed by a reduction in the cartilage area in the posterior region at twelve weeks. Superimposed imaging and histomorphometrical analysis of the condyle revealed that BMP signaling primarily affects the posterior part of the condyle, while mastication affects the anterior part. CONCLUSIONS Using 3D landmark-based geometric morphometrics and histological assessments of the mandible, we demonstrated that BMP signaling and mechanical loading reciprocally contribute to the morphological alterations of the mandible and condyle during postnatal development.
Collapse
Affiliation(s)
- Amber Uptegrove
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Coral Chen
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Madison Sahagun-Bisson
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Anshul K Kulkarni
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Ke'ale W Louie
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Hiroki Ueharu
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA.
| | - Maiko Omi-Sugihara
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA; Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan.
| |
Collapse
|
2
|
Bertels JC, He G, Long F. Metabolic reprogramming in skeletal cell differentiation. Bone Res 2024; 12:57. [PMID: 39394187 PMCID: PMC11470040 DOI: 10.1038/s41413-024-00374-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/13/2024] Open
Abstract
The human skeleton is a multifunctional organ made up of multiple cell types working in concert to maintain bone and mineral homeostasis and to perform critical mechanical and endocrine functions. From the beginning steps of chondrogenesis that prefigures most of the skeleton, to the rapid bone accrual during skeletal growth, followed by bone remodeling of the mature skeleton, cell differentiation is integral to skeletal health. While growth factors and nuclear proteins that influence skeletal cell differentiation have been extensively studied, the role of cellular metabolism is just beginning to be uncovered. Besides energy production, metabolic pathways have been shown to exert epigenetic regulation via key metabolites to influence cell fate in both cancerous and normal tissues. In this review, we will assess the role of growth factors and transcription factors in reprogramming cellular metabolism to meet the energetic and biosynthetic needs of chondrocytes, osteoblasts, or osteoclasts. We will also summarize the emerging evidence linking metabolic changes to epigenetic modifications during skeletal cell differentiation.
Collapse
Affiliation(s)
- Joshua C Bertels
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Guangxu He
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Orthopedics, The Second Xiangya Hospital, Changsha, Hunan, China
| | - Fanxin Long
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Umur E, Bulut SB, Yiğit P, Bayrak E, Arkan Y, Arslan F, Baysoy E, Kaleli-Can G, Ayan B. Exploring the Role of Hormones and Cytokines in Osteoporosis Development. Biomedicines 2024; 12:1830. [PMID: 39200293 PMCID: PMC11351445 DOI: 10.3390/biomedicines12081830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
The disease of osteoporosis is characterized by impaired bone structure and an increased risk of fractures. There is a significant impact of cytokines and hormones on bone homeostasis and the diagnosis of osteoporosis. As defined by the World Health Organization (WHO), osteoporosis is defined as having a bone mineral density (BMD) that is 2.5 standard deviations (SD) or more below the average for young and healthy women (T score < -2.5 SD). Cytokines and hormones, particularly in the remodeling of bone between osteoclasts and osteoblasts, control the differentiation and activation of bone cells through cytokine networks and signaling pathways like the nuclear factor kappa-B ligand (RANKL)/the receptor of RANKL (RANK)/osteoprotegerin (OPG) axis, while estrogen, parathyroid hormones, testosterone, and calcitonin influence bone density and play significant roles in the treatment of osteoporosis. This review aims to examine the roles of cytokines and hormones in the pathophysiology of osteoporosis, evaluating current diagnostic methods, and highlighting new technologies that could help for early detection and treatment of osteoporosis.
Collapse
Affiliation(s)
- Egemen Umur
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Safiye Betül Bulut
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Pelin Yiğit
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Emirhan Bayrak
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Yaren Arkan
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Fahriye Arslan
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Engin Baysoy
- Department of Biomedical Engineering, Bahçeşehir University, İstanbul 34353, Türkiye
| | - Gizem Kaleli-Can
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Bugra Ayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Varma S, Molangiri A, Mudavath S, Ananthan R, Rajanna A, Duttaroy AK, Basak S. Exposure to BPA and BPS during pregnancy disrupts the bone mineralization in the offspring. Food Chem Toxicol 2024; 189:114772. [PMID: 38821392 DOI: 10.1016/j.fct.2024.114772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/03/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Exposure to plastic-derived estrogen-mimicking endocrine-disrupting bisphenols can have a long-lasting effect on bone health. However, gestational exposure to bisphenol A (BPA) and its analogue, bisphenol S (BPS), on offspring's bone mineralization is unclear. The effects of in-utero bisphenol exposure were examined on the offspring's bone parameters. BPA and BPS (0.0, 0.4 μg/kg bw) were administered to pregnant Wistar rats via oral gavage from gestational day 4-21. Maternal exposure to BPA and BPS increased bone mineral content and density in the offspring aged 30 and 90 days (P < 0.05). Plasma analysis revealed that alkaline phosphatase, and Gla-type osteocalcin were significantly elevated in the BPS-exposed offspring (P < 0.05). The expression of BMP1, BMP4, and their signaling mediators SMAD1 mRNAs were decreased in BPS-exposed osteoblast SaOS-2 cells (P < 0.05). The expression of extracellular matrix proteins such as ALPL, COL1A1, DMP1, and FN1 were downregulated (P < 0.05). Bisphenol co-incubation with noggin decreased TGF-β1 expression, indicating its involvement in bone mineralization. Altered mineralization could be due to dysregulated expression of bone morphogenetic proteins and signalling mediators in the osteoblast cells. Thus, bisphenol exposure during gestation altered growth and bone mineralization in the offspring, possibly by modulating the expression of Smad-dependent BMP/TGF-β1 signalling mediators.
Collapse
Affiliation(s)
- Saikanth Varma
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Archana Molangiri
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Sreedhar Mudavath
- Food Chemistry Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Rajendran Ananthan
- Food Chemistry Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Ajumeera Rajanna
- Cell Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India.
| |
Collapse
|
5
|
Lademann F, Rijntjes E, Köhrle J, Tsourdi E, Hofbauer LC, Rauner M. Hyperthyroidism-driven bone loss depends on BMP receptor Bmpr1a expression in osteoblasts. Commun Biol 2024; 7:548. [PMID: 38719881 PMCID: PMC11078941 DOI: 10.1038/s42003-024-06227-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
Hyperthyroidism is a well-known trigger of high bone turnover that can lead to the development of secondary osteoporosis. Previously, we have shown that blocking bone morphogenetic protein (BMP) signaling systemically with BMPR1A-Fc can prevent bone loss in hyperthyroid mice. To distinguish between bone cell type-specific effects, conditional knockout mice lacking Bmpr1a in either osteoclast precursors (LysM-Cre) or osteoprogenitors (Osx-Cre) were rendered hyperthyroid and their bone microarchitecture, strength and turnover were analyzed. While hyperthyroidism in osteoclast precursor-specific Bmpr1a knockout mice accelerated bone resorption leading to bone loss just as in wildtype mice, osteoprogenitor-specific Bmpr1a deletion prevented an increase of bone resorption and thus osteoporosis with hyperthyroidism. In vitro, wildtype but not Bmpr1a-deficient osteoblasts responded to thyroid hormone (TH) treatment with increased differentiation and activity. Furthermore, we found an elevated Rankl/Opg ratio with TH excess in osteoblasts and bone tissue from wildtype mice, but not in Bmpr1a knockouts. In line, expression of osteoclast marker genes increased when osteoclasts were treated with supernatants from TH-stimulated wildtype osteoblasts, in contrast to Bmpr1a-deficient cells. In conclusion, we identified the osteoblastic BMP receptor BMPR1A as a main driver of osteoporosis in hyperthyroid mice promoting TH-induced osteoblast activity and potentially its coupling to high osteoclastic resorption.
Collapse
Affiliation(s)
- Franziska Lademann
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Eddy Rijntjes
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Josef Köhrle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Elena Tsourdi
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany.
| |
Collapse
|
6
|
Wu M, Wu S, Chen W, Li YP. The roles and regulatory mechanisms of TGF-β and BMP signaling in bone and cartilage development, homeostasis and disease. Cell Res 2024; 34:101-123. [PMID: 38267638 PMCID: PMC10837209 DOI: 10.1038/s41422-023-00918-9] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Transforming growth factor-βs (TGF-βs) and bone morphometric proteins (BMPs) belong to the TGF-β superfamily and perform essential functions during osteoblast and chondrocyte lineage commitment and differentiation, skeletal development, and homeostasis. TGF-βs and BMPs transduce signals through SMAD-dependent and -independent pathways; specifically, they recruit different receptor heterotetramers and R-Smad complexes, resulting in unique biological readouts. BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner. BMPs and TGF-β have opposite functions in articular cartilage homeostasis. Moreover, TGF-β has a specific role in maintaining the osteocyte network. The precise activation of BMP and TGF-β signaling requires regulatory machinery at multiple levels, including latency control in the matrix, extracellular antagonists, ubiquitination and phosphorylation in the cytoplasm, nucleus-cytoplasm transportation, and transcriptional co-regulation in the nuclei. This review weaves the background information with the latest advances in the signaling facilitated by TGF-βs and BMPs, and the advanced understanding of their diverse physiological functions and regulations. This review also summarizes the human diseases and mouse models associated with disordered TGF-β and BMP signaling. A more precise understanding of the BMP and TGF-β signaling could facilitate the development of bona fide clinical applications in treating bone and cartilage disorders.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
7
|
Al Maruf DSA, Xin H, Cheng K, Garcia AG, Mohseni-Dargah M, Ben-Sefer E, Tomaskovic-Crook E, Crook JM, Clark JR. Bioengineered cartilaginous grafts for repairing segmental mandibular defects. J Tissue Eng 2024; 15. [DOI: 10.1177/20417314241267017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Reconstructing critical-sized craniofacial bone defects is a global healthcare challenge. Current methods, like autologous bone transplantation, face limitations. Bone tissue engineering offers an alternative to autologous bone, with traditional approaches focusing on stimulating osteogenesis via the intramembranous ossification (IMO) pathway. However, IMO falls short in addressing larger defects, particularly in clinical scenarios where there is insufficient vascularisation. This review explores redirecting bone regeneration through endochondral ossification (ECO), a process observed in long bone healing stimulated by hypoxic conditions. Despite its promise, gaps exist in applying ECO to bone tissue engineering experiments, requiring the elucidation of key aspects such as cell sources, biomaterials and priming protocols. This review discusses various scaffold biomaterials and cellular sources for chondrogenesis and hypertrophic chondrocyte priming, mirroring the ECO pathway. The review highlights challenges in current endochondral priming and proposes alternative approaches. Emphasis is on segmental mandibular defect repair, offering insights for future research and clinical application. This concise review aims to advance bone tissue engineering by addressing critical gaps in ECO strategies.
Collapse
Affiliation(s)
- D S Abdullah Al Maruf
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Hai Xin
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Kai Cheng
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| | - Alejandro Garcia Garcia
- Cell, Tissue and Organ Engineering Laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
| | - Masoud Mohseni-Dargah
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
| | - Eitan Ben-Sefer
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Eva Tomaskovic-Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jeremy Micah Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jonathan Robert Clark
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| |
Collapse
|
8
|
Richardson L, Wilcockson SG, Guglielmi L, Hill CS. Context-dependent TGFβ family signalling in cell fate regulation. Nat Rev Mol Cell Biol 2023; 24:876-894. [PMID: 37596501 DOI: 10.1038/s41580-023-00638-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/20/2023]
Abstract
The transforming growth factor-β (TGFβ) family are a large group of evolutionarily conserved cytokines whose signalling modulates cell fate decision-making across varying cellular contexts at different stages of life. Here we discuss new findings in early embryos that reveal how, in contrast to our original understanding of morphogen interpretation, robust cell fate specification can originate from a noisy combination of signalling inputs and a broad range of signalling levels. We compare this evidence with novel findings on the roles of TGFβ family signalling in tissue maintenance and homeostasis during juvenile and adult life, spanning the skeletal, haemopoietic and immune systems. From these comparisons, it emerges that in contrast to robust developing systems, relatively small perturbations in TGFβ family signalling have detrimental effects at later stages in life, leading to aberrant cell fate specification and disease, for example in cancer or congenital disorders. Finally, we highlight novel strategies to target and amend dysfunction in signalling and discuss how gleaning knowledge from different fields of biology can help in the development of therapeutics for aberrant TGFβ family signalling in disease.
Collapse
Affiliation(s)
- Louise Richardson
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
| | - Scott G Wilcockson
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
| | - Luca Guglielmi
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
9
|
Ruan X, Gu J, Chen M, Zhao F, Aili M, Zhang D. Multiple roles of ALK3 in osteoarthritis. Bone Joint Res 2023; 12:397-411. [PMID: 37394235 PMCID: PMC10315222 DOI: 10.1302/2046-3758.127.bjr-2022-0310.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by progressive cartilage degradation, synovial membrane inflammation, osteophyte formation, and subchondral bone sclerosis. Pathological changes in cartilage and subchondral bone are the main processes in OA. In recent decades, many studies have demonstrated that activin-like kinase 3 (ALK3), a bone morphogenetic protein receptor, is essential for cartilage formation, osteogenesis, and postnatal skeletal development. Although the role of bone morphogenetic protein (BMP) signalling in articular cartilage and bone has been extensively studied, many new discoveries have been made in recent years around ALK3 targets in articular cartilage, subchondral bone, and the interaction between the two, broadening the original knowledge of the relationship between ALK3 and OA. In this review, we focus on the roles of ALK3 in OA, including cartilage and subchondral bone and related cells. It may be helpful to seek more efficient drugs or treatments for OA based on ALK3 signalling in future.
Collapse
Affiliation(s)
- Xianchun Ruan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinning Gu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Mingyang Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fulin Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Munire Aili
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Ruan X, Zhang Z, Aili M, Luo X, Wei Q, Zhang D, Bai M. Activin receptor-like kinase 3: a critical modulator of development and function of mineralized tissues. Front Cell Dev Biol 2023; 11:1209817. [PMID: 37457289 PMCID: PMC10347416 DOI: 10.3389/fcell.2023.1209817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Mineralized tissues, such as teeth and bones, pose significant challenges for repair due to their hardness, low permeability, and limited blood flow compared to soft tissues. Bone morphogenetic proteins (BMPs) have been identified as playing a crucial role in mineralized tissue formation and repair. However, the application of large amounts of exogenous BMPs may cause side effects such as inflammation. Therefore, it is necessary to identify a more precise molecular target downstream of the ligands. Activin receptor-like kinase 3 (ALK3), a key transmembrane receptor, serves as a vital gateway for the transmission of BMP signals, triggering cellular responses. Recent research has yielded new insights into the regulatory roles of ALK3 in mineralized tissues. Experimental knockout or mutation of ALK3 has been shown to result in skeletal dysmorphisms and failure of tooth formation, eruption, and orthodontic tooth movement. This review summarizes the roles of ALK3 in mineralized tissue regulation and elucidates how ALK3-mediated signaling influences the physiology and pathology of teeth and bones. Additionally, this review provides a reference for recommended basic research and potential future treatment strategies for the repair and regeneration of mineralized tissues.
Collapse
Affiliation(s)
- Xianchun Ruan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhaowei Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Munire Aili
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiang Luo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Qiang Wei
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Mingru Bai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Omi M, Koneru T, Lyu Y, Haraguchi A, Kamiya N, Mishina Y. Increased BMP-Smad signaling does not affect net bone mass in long bones. Front Physiol 2023; 14:1145763. [PMID: 37064883 PMCID: PMC10101206 DOI: 10.3389/fphys.2023.1145763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) have been used for orthopedic and dental application due to their osteoinductive properties; however, substantial numbers of adverse reactions such as heterotopic bone formation, increased bone resorption and greater cancer risk have been reported. Since bone morphogenetic proteins signaling exerts pleiotropic effects on various tissues, it is crucial to understand tissue-specific and context-dependent functions of bone morphogenetic proteins. We previously reported that loss-of-function of bone morphogenetic proteins receptor type IA (BMPR1A) in osteoblasts leads to more bone mass in mice partly due to inhibition of bone resorption, indicating that bone morphogenetic protein signaling in osteoblasts promotes osteoclast function. On the other hand, hemizygous constitutively active (ca) mutations for BMPR1A (caBmpr1awt/+) in osteoblasts result in higher bone morphogenetic protein signaling activity and no overt skeletal changes in adult mice. Here, we further bred mice for heterozygous null for Bmpr1a (Bmpr1a+/−) and homozygous mutations of caBmpr1a (caBmpr1a+/+) crossed with Osterix-Cre transgenic mice to understand how differences in the levels of bone morphogenetic protein signaling activity specifically in osteoblasts contribute to bone phenotype. We found that Bmpr1a+/−, caBmpr1awt/+ and caBmpr1a+/+ mice at 3 months of age showed no overt bone phenotypes in tibiae compared to controls by micro-CT and histological analysis although BMP-Smad signaling is increased in both caBmpr1awt/+ and caBmpr1a+/+ tibiae and decreased in the Bmpr1a+/− mice compared to controls. Gene expression analysis demonstrated that slightly higher levels of bone formation markers and resorption markers along with levels of bone morphogenetic protein-Smad signaling, however, there was no significant changes in TRAP positive cells in tibiae. These findings suggest that changes in bone morphogenetic protein signaling activity within differentiating osteoblasts does not affect net bone mass in the adult stage, providing insights into the concerns in the clinical setting such as high-dose and unexpected side effects of bone morphogenetic protein application.
Collapse
Affiliation(s)
- Maiko Omi
- Department of Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Tejaswi Koneru
- Department of Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Yishan Lyu
- Department of Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Ai Haraguchi
- Department of Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Nobuhiro Kamiya
- Department of Budo and Sport Studies, Faculty of Budo and Sport Studies, Tenri University, Nara, Japan
| | - Yuji Mishina
- Department of Biologic and Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
- *Correspondence: Yuji Mishina,
| |
Collapse
|
12
|
Wang M, Fan J, Wang A, Jin X, Zhang Z, Hu X, Liu L, Zhao Y, Li Y. Effect of local application of bone morphogenetic protein -2 on experimental tooth movement and biological remodeling in rats. Front Physiol 2023; 14:1111857. [PMID: 37143931 PMCID: PMC10151543 DOI: 10.3389/fphys.2023.1111857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Background: This study attempts to detect the potential effects of local bone morphogenetic protein -2 (BMP-2) on orthodontic tooth movement and periodontal tissue remodeling. Methods: Forty adult SD rats were randomly divided into four groups: blank control group, unilateral injection of BMP-2 on the pressure side or tension side of orthodontic teeth and bilateral injection of BMP-2. Their maxillary first molar was moved by a 30 g constant force closed coil spring. 60 μL of BMP-2 with a concentration of 0.5 μg/mL was injected into each part at a time. In addition, three rats were selected as healthy control rats without any intervention. Fluorescent labeled BMP-2 was used to observe the distribution of exogenous BMP-2 in tissues. Micro-CT was used to measure the microscopic parameters of tooth displacement, trabecular bone and root absorption volume. Three different histological methods were used to observe the changes of tissue remodeling, and then the number of osteoclasts and the content of collagen fibers were calculated. Results: Compared with the blank control group, BMP-2 injection reduced the movement distance and increased the collagen fiber content and bone mass (p < 0.01). There was no significant difference in tooth movement distance, BV/TV ratio and BMD between injection sites in unilateral injection group (p > 0.05). In the case of bilateral injection of BMP-2, the osteogenesis is enhanced. Unilateral injection of BMP-2 did not promote root resorption, but double injection showed root resorption (p < 0.01). Conclusion: Our study does show that the osteogenesis of BMP-2 is dose-dependent rather than site-dependent when a certain amount of BMP-2 is applied around orthodontic teeth. Local application of BMP-2 around orthodontic teeth in an appropriate way can enhance bone mass and tooth anchorage without increasing the risk of root absorption volume. However, high levels of BMP-2 may cause aggressive root resorption. These findings are of great significance, that is, BMP-2 is an effective target for regulating orthodontic tooth movement.
Collapse
Affiliation(s)
- Menglin Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jiadong Fan
- Department of Stomatology, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Aoao Wang
- Senior Department of Orthopedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiang Jin
- Department of Stomatology, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhenbao Zhang
- Department of Stomatology, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiantong Hu
- Senior Department of Orthopedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Le Liu
- Department of Stomatology, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Le Liu, ; Yantao Zhao, ; Yanfeng Li,
| | - Yantao Zhao
- Department of Stomatology, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
- Senior Department of Orthopedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
- Beijing Engineering Research Center of Orthopedics Implants, Beijing, China
- State Key Laboratory of Military Stomatology, Xi’an, China
- *Correspondence: Le Liu, ; Yantao Zhao, ; Yanfeng Li,
| | - Yanfeng Li
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Le Liu, ; Yantao Zhao, ; Yanfeng Li,
| |
Collapse
|
13
|
Rosyida NF, Ana ID, Alhasyimi AA. The Use of Polymers to Enhance Post-Orthodontic Tooth Stability. Polymers (Basel) 2022; 15:polym15010103. [PMID: 36616453 PMCID: PMC9824751 DOI: 10.3390/polym15010103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/11/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Relapse after orthodontic treatment occurs at a rate of about 70 to 90%, and this phenomenon is an orthodontic issue that has not yet been resolved. Retention devices are one attempt at prevention, but they require a considerable amount of time. Most orthodontists continue to find it challenging to manage orthodontic relapse; therefore, additional research is required. In line with existing knowledge regarding the biological basis of relapse, biomedical engineering approaches to relapse regulation show promise. With so many possible uses in biomedical engineering, polymeric materials have long been at the forefront of the materials world. Orthodontics is an emerging field, and scientists are paying a great deal of attention to polymers because of their potential applications in this area. In recent years, the controlled release of bisphosphonate risedronate using a topically applied gelatin hydrogel has been demonstrated to be effective in reducing relapse. Simvastatin encapsulation in exosomes generated from periodontal ligament stem cells can promote simvastatin solubility and increase the inhibitory action of orthodontic relapse. Moreover, the local injection of epigallocatechin gallate-modified gelatin suppresses osteoclastogenesis and could be developed as a novel treatment method to modify tooth movement and inhibit orthodontic relapse. Furthermore, the intrasulcular administration of hydrogel carbonated hydroxyapatite-incorporated advanced platelet-rich fibrin has been shown to minimize orthodontic relapse. The objective of this review was to provide an overview of the use of polymer materials to reduce post-orthodontic relapse. We assume that bone remodeling is a crucial factor even though the exact process by which orthodontic correction is lost after retention is not fully known. Delivery of a polymer containing elements that altered osteoclast activity inhibited osteoclastogenesis and blocking orthodontic relapse. The most promising polymeric materials and their potential orthodontic uses for the prevention of orthodontic relapse are also discussed.
Collapse
Affiliation(s)
- Niswati Fathmah Rosyida
- Department of Orthodontics, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research and Innovation Agency (BRIN), Jakarta 10340, Indonesia
| | - Ananto Ali Alhasyimi
- Department of Orthodontics, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Correspondence: ; Tel.: +62-82136708250
| |
Collapse
|
14
|
Zhang Y, Zhao Y, Xie Z, Li M, Liu Y, Tu X. Activating Wnt/β-Catenin Signaling in Osteocytes Promotes Osteogenic Differentiation of BMSCs through BMP-7. Int J Mol Sci 2022; 23:ijms232416045. [PMID: 36555684 PMCID: PMC9785209 DOI: 10.3390/ijms232416045] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Bone formation is critically needed in orthopedic clinical practice. We found that, bone morphogenetic protein-7 (BMP-7) gene expression was significantly increased in fractured mice, which activates canonical Wnt signaling exclusively in osteocytes. Wnt and BMP signaling appear to exhibit synergistic or antagonistic effects in different kinds of cells. However, the communication between Wnt/β-catenin signaling and BMP signaling in osteocytes is almost unknown. Our study verified in vitro that BMP-7 expression was significantly increased when Wnt signaling was activated in osteocytes. Next, BMP-7 in osteocytes was overexpressed using an adenovirus, the osteogenesis of bone marrow stem cells (BMSCs) was enhanced, when cocultured with osteocytes. On the contrary, BMP-7 in osteocytes was silenced using an adenovirus, the osteogenesis of bone marrow stem cells (BMSCs) was weakened. In addition, the osteogenesis of BMSCs was no longer promoted by Wnt-activated osteocytes when BMP-7 was silenced. Therefore, the results showed that BMP-7 mediated the anabolic actions of Wnt/β-catenin signaling in osteocytes. Our study provides new evidence for the clinical application of BMP-7-overexpressed osteocytes.
Collapse
Affiliation(s)
- Yining Zhang
- Correspondence: (Y.Z.); (X.T.); Tel.: +86-23-6365-1934 (X.T.)
| | | | | | | | | | - Xiaolin Tu
- Correspondence: (Y.Z.); (X.T.); Tel.: +86-23-6365-1934 (X.T.)
| |
Collapse
|
15
|
Zuardi LR, de Oliveira FS, Fernandes RR, Gomes MPO, Spriano S, Nanci A, de Oliveira PT. Effects of rmBMP-7 on Osteoblastic Cells Grown on a Nanostructured Titanium Surface. Biomimetics (Basel) 2022; 7:biomimetics7030136. [PMID: 36134940 PMCID: PMC9496167 DOI: 10.3390/biomimetics7030136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/06/2022] [Accepted: 09/14/2022] [Indexed: 01/23/2023] Open
Abstract
This study evaluates the effects of the availability of exogenous BMP-7 on osteoblastic cells’ differentiation on a nanotextured Ti surface obtained by chemical etching (Nano-Ti). The MC3T3-E1 and UMR-106 osteoblastic cell lines were cultured for 5 and 7 days, respectively, on a Nano-Ti surface and on a control surface (Control-Ti) in an osteogenic medium supplemented with either 40 or 200 ng/mL recombinant mouse (rm) BMP-7. The results showed that MC3T3-E1 cells exhibited distinct responsiveness when exposed to each of the two rmBMP-7 concentrations, irrespective of the surface. Even with 40 ng/mL rmBMP-7, important osteogenic effects were noticed for Control-Ti in terms of cell proliferation potential; Runx2, Osx, Alp, Bsp, Opn, and Smad1 mRNA expression; and in situ ALP activity. For Nano-Ti, the effects were limited to higher Alp, Bsp, and Opn mRNA expression and in situ ALP activity. On both surfaces, the osteogenic potential of UMR-106 cultures remained unaltered with 40 ng/mL rmBMP-7, but it was significantly reduced when the cultures were exposed to the 200 ng/mL concentration. The availability of rmBMP-7 to pre-osteoblastic cells at the concentrations used alters the expression profile of osteoblast markers, indicative of the acquisition of a more advanced stage of osteoblastic differentiation. This occurs less pronouncedly on the nanotextured Ti and without reflecting in higher mineralized matrix production by differentiated osteoblasts on both surfaces.
Collapse
Affiliation(s)
- Leonardo Raphael Zuardi
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Fabíola Singaretti de Oliveira
- Department of Oral and Maxillofacial Surgery and Periodontics, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Roger Rodrigo Fernandes
- Department of Oral and Maxillofacial Surgery and Periodontics, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Maria Paula Oliveira Gomes
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Silvia Spriano
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Torino, Italy
| | - Antonio Nanci
- Faculté de Médecine Dentaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Paulo Tambasco de Oliveira
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
- Correspondence: ; Tel.: +55-16-99623-3663
| |
Collapse
|
16
|
Park KR, Park JI, Lee S, Yoo K, Kweon GR, Kwon IK, Yun HM, Hong JT. Chi3L1 is a therapeutic target in bone metabolism and a potential clinical marker in patients with osteoporosis. Pharmacol Res 2022; 184:106423. [PMID: 36064078 DOI: 10.1016/j.phrs.2022.106423] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/20/2022] [Accepted: 08/30/2022] [Indexed: 01/03/2023]
Abstract
BMP2 is clinically used as an ectopic bone inducer and plays a significant role in bone development, formation, and diseases. Chitinase 3-like 1 protein (Chi3L1) is found in the skeletal system. However, Chi3L1-mediated bone metabolism and aging-related bone erosion via BMP2 signaling have not yet been demonstrated. Herein, Chi3L1 increased BMP2-induced osteoblast differentiation in mesenchymal precursor cells and human primary osteoblasts. Chi3L1KO(-/-) showed abnormal bone development, and primary osteoblasts isolated from Chi3L1KO(-/-) exhibited impaired osteoblast differentiation and maturation. Chi3L1 also potentiated BMP2 signaling and RUNX2 expression in primary osteoblasts. Chi3L1 interacted with BMPRIa, which increased the surface expression of BMPRIa and promoted BMP2 signaling to induce osteoblast differentiation. Chi3L1KO(-/-) mice showed bone formation reduced with a decrease in RUNX2 expression in calvarial defects. Chi3L1KO(-/-) mice exhibited aging-related osteoporotic bone loss with decreases in the levels of RUNX2 and OPG, while serum PYD level and osteoclast number increased. Chi3L1 increased OPG via non-canonical BMP2 signaling in osteoblasts, which suppressed osteoclastogenesis in BMMs. Furthermore, ROC analysis showed that serum Chi3L1 level clinically decreased in osteoporosis patients. Our findings demonstrate that Chi3L1 promotes bone formation, suppresses osteoclastogenesis, and prevents aging-related osteoporosis.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea.
| | - Jae-Il Park
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61751, Republic of Korea.
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea.
| | - Kyeongwon Yoo
- KRIBB/Bio-venture Center, Daejeon 34141, Republic of Korea.
| | - Gi-Ryang Kweon
- Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Chungnam 34134, Republic of Korea.
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02453, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
17
|
Perforated Hydrogels Consisting of Cholesterol-Bearing Pullulan (CHP) Nanogels: A Newly Designed Scaffold for Bone Regeneration Induced by RANKL-Binding Peptides and BMP-2. Int J Mol Sci 2022; 23:ijms23147768. [PMID: 35887115 PMCID: PMC9316061 DOI: 10.3390/ijms23147768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
The receptor activator of NF-κB ligand (RANKL)-binding peptide, OP3-4, is known to stimulate bone morphogenetic protein (BMP)-2-induced bone formation, but peptides tend to aggregate and lose their bioactivity. Cholesterol-bearing pullulan (CHP) nanogel scaffold has been shown to prevent aggregation of peptides and to allow their sustained release and activity; however, the appropriate design of CHP nanogels to conduct local bone formation needs to be developed. In the present study, we investigated the osteoconductive capacity of a newly synthesized CHP nanogel, CHPA using OP3-4 and BMP-2. We also clarified the difference between perforated and nonperforated CHPA impregnated with the two signaling molecules. Thirty-six, five-week-old male BALB/c mice were used for the calvarial defect model. The mice were euthanized at 6 weeks postoperatively. A higher cortical bone mineral content and bone formation rate were observed in the perforated scaffold in comparison to the nonperforated scaffold, especially in the OP3-4/BMP-2 combination group. The degradation rate of scaffold material in the perforated OP3-4/BMP-2 combination group was lower than that in the nonperforated group. These data suggest that perforated CHPA nanogel could lead to local bone formation induced by OP3-4 and BMP–2 and clarified the appropriate degradation rate for inducing local bone formation when CHPA nanogels are designed to be perforated.
Collapse
|
18
|
Zou N, Liu R, Li C. Cathepsin K+ Non-Osteoclast Cells in the Skeletal System: Function, Models, Identity, and Therapeutic Implications. Front Cell Dev Biol 2022; 10:818462. [PMID: 35912093 PMCID: PMC9326176 DOI: 10.3389/fcell.2022.818462] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cathepsin K (Ctsk) is a cysteine protease of the papain superfamily initially identified in differentiated osteoclasts; it plays a critical role in degrading the bone matrix. However, subsequent in vivo and in vitro studies based on animal models elucidate novel subpopulations of Ctsk-expressing cells, which display markers and properties of mesenchymal stem/progenitor cells. This review introduces the function, identity, and role of Ctsk+ cells and their therapeutic implications in related preclinical osseous disorder models. It also summarizes the available in vivo models for studying Ctsk+ cells and their progeny. Further investigations of detailed properties and mechanisms of Ctsk+ cells in transgenic models are required to guide potential therapeutic targets in multiple diseases in the future.
Collapse
Affiliation(s)
- Nanyu Zou
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Ran Liu
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- *Correspondence: Changjun Li,
| |
Collapse
|
19
|
Xie X, Xu C, Zhao L, Wu Y, Feng JQ, Wang J. Axin2-expressing cells in the PDL are regulated by BMP signaling and play a pivotal role in periodontium development. J Clin Periodontol 2022; 49:945-956. [PMID: 35634660 DOI: 10.1111/jcpe.13666] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/05/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND To date, controversies still exist regarding the exact cellular origin and regulatory mechanisms of periodontium development, which hinders efforts to achieve ideal periodontal tissue regeneration. Axin2-expressing cells in the periodontal ligament (PDL) have been shown to be a novel progenitor cell population that is essential for periodontal homeostasis. In the current study, we aimed to elucidate the regulatory role of bone morphogenetic protein receptor type 1A (BMPR1A)-mediated BMP signaling in Axin2-expressing cells during periodontium development. METHODS Two strains of Axin2 gene reporter mice, Axin2lacZ/+ and Axin2CreERT2/+ ; R26RtdTomato/+ mice, were used. We next generated Axin2CreERT2/+ ; R26RDTA/+ ; R26RtdTomato/+ mice to genetically ablate of Axin2-lineage cells. Axin2CreERT2/+ ; Bmpr1afl/fl ; R26RtdTomato/+ mice were established to conditionally knock out Bmpr1a in Axin2-lineage cells. Multiple approaches, including micro-CT, calcein green and alizarin red double-labeling, scanning electron microscopy, and histological and immunostaining assays, were used to analyze periodontal phenotypes and molecular mechanisms. RESULTS X-gal staining revealed that Axin2-expressing cells in the PDL were mainly distributed along the alveolar bone and cementum surface. Cell lineage tracing and cell ablation assays further demonstrated the indispensable role of Axin2-expressing cells in periodontium development. Next, we found that conditional knockout of Bmpr1a in Axin2-lineage cells led to periodontal defects, which were characterized by alveolar bone loss, impaired cementogenesis, and abnormal Sharpey's fibers. CONCLUSIONS Our findings suggest that Axin2-expressing cells in the PDL are essential for periodontium development, which is regulated by BMP signaling. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xudong Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chunmei Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
The Emerging Role of Cell Transdifferentiation in Skeletal Development and Diseases. Int J Mol Sci 2022; 23:ijms23115974. [PMID: 35682655 PMCID: PMC9180549 DOI: 10.3390/ijms23115974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The vertebrate musculoskeletal system is known to be formed by mesenchymal stem cells condensing into tissue elements, which then differentiate into cartilage, bone, tendon/ligament, and muscle cells. These lineage-committed cells mature into end-stage differentiated cells, like hypertrophic chondrocytes and osteocytes, which are expected to expire and to be replaced by newly differentiated cells arising from the same lineage pathway. However, there is emerging evidence of the role of cell transdifferentiation in bone development and disease. Although the concept of cell transdifferentiation is not new, a breakthrough in cell lineage tracing allowed scientists to trace cell fates in vivo. Using this powerful tool, new theories have been established: (1) hypertrophic chondrocytes can transdifferentiate into bone cells during endochondral bone formation, fracture repair, and some bone diseases, and (2) tendon cells, beyond their conventional role in joint movement, directly participate in normal bone and cartilage formation, and ectopic ossification. The goal of this review is to obtain a better understanding of the key roles of cell transdifferentiation in skeletal development and diseases. We will first review the transdifferentiation of chondrocytes to bone cells during endochondral bone formation. Specifically, we will include the history of the debate on the fate of chondrocytes during bone formation, the key findings obtained in recent years on the critical factors and molecules that regulate this cell fate change, and the role of chondrocyte transdifferentiation in skeletal trauma and diseases. In addition, we will also summarize the latest discoveries on the novel roles of tendon cells and adipocytes on skeletal formation and diseases.
Collapse
|
21
|
Bordukalo-Nikšić T, Kufner V, Vukičević S. The Role Of BMPs in the Regulation of Osteoclasts Resorption and Bone Remodeling: From Experimental Models to Clinical Applications. Front Immunol 2022; 13:869422. [PMID: 35558080 PMCID: PMC9086899 DOI: 10.3389/fimmu.2022.869422] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
In response to mechanical forces and the aging process, bone in the adult skeleton is continuously remodeled by a process in which old and damaged bone is removed by bone-resorbing osteoclasts and subsequently is replaced by new bone by bone-forming cells, osteoblasts. During this essential process of bone remodeling, osteoclastic resorption is tightly coupled to osteoblastic bone formation. Bone-resorbing cells, multinuclear giant osteoclasts, derive from the monocyte/macrophage hematopoietic lineage and their differentiation is driven by distinct signaling molecules and transcription factors. Critical factors for this process are Macrophage Colony Stimulating Factor (M-CSF) and Receptor Activator Nuclear Factor-κB Ligand (RANKL). Besides their resorption activity, osteoclasts secrete coupling factors which promote recruitment of osteoblast precursors to the bone surface, regulating thus the whole process of bone remodeling. Bone morphogenetic proteins (BMPs), a family of multi-functional growth factors involved in numerous molecular and signaling pathways, have significant role in osteoblast-osteoclast communication and significantly impact bone remodeling. It is well known that BMPs help to maintain healthy bone by stimulating osteoblast mineralization, differentiation and survival. Recently, increasing evidence indicates that BMPs not only help in the anabolic part of bone remodeling process but also significantly influence bone catabolism. The deletion of the BMP receptor type 1A (BMPRIA) in osteoclasts increased osteoblastic bone formation, suggesting that BMPR1A signaling in osteoclasts regulates coupling to osteoblasts by reducing bone-formation activity during bone remodeling. The dual effect of BMPs on bone mineralization and resorption highlights the essential role of BMP signaling in bone homeostasis and they also appear to be involved in pathological processes in inflammatory disorders affecting bones and joints. Certain BMPs (BMP2 and -7) were approved for clinical use; however, increased bone resorption rather than formation were observed in clinical applications, suggesting the role BMPs have in osteoclast activation and subsequent osteolysis. Here, we summarize the current knowledge of BMP signaling in osteoclasts, its role in osteoclast resorption, bone remodeling, and osteoblast–osteoclast coupling. Furthermore, discussion of clinical application of recombinant BMP therapy is based on recent preclinical and clinical studies.
Collapse
Affiliation(s)
- Tatjana Bordukalo-Nikšić
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vera Kufner
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Slobodan Vukičević
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
22
|
王 欣, 牛 志, 张 德, 谢 静, 周 学. [Latest Research Findings on the Regulation of Bone Homeostasis by ALK3]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2022; 53:517-522. [PMID: 35642164 PMCID: PMC10409440 DOI: 10.12182/20220560304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Indexed: 06/15/2023]
Abstract
Bone remodeling, which is well orchestrated by osteogenesis of osteoblasts and osteoclastogenesis of osteoclasts, maintains the homeostasis of osteal development and metabolism under physiological conditions. Bone morphogenetic protein receptor type 1A, also known as activin receptor-like kinase 3 (ALK3), which exists on cytomembrane, is one of the key receptors of BMP factors, and is an important "gateway" that regulates the entrance of BMP signaling into cells in order to perform biological functions. The roles of BMP signaling in bone remodeling have been extensively studied. Many new discoveries have been reported in recent years through research based on transgenic mice models and focused on ALK3 as targets, shedding new light on the regulations of bone remodeling, cartilage and joint development, and the occurrence and treatment of bone-related diseases. Established understanding has been expanded, but new challenges on existing clinical application of BMPs also appeared. Hence, we reviewed recent studies on ALK3's involvement in bone formation and bone resorption, analyzed its mechanism of action in bone regulation, summarized the roles of ALK3 in the development of cartilage and temporomandibular joint, and reported the latest progress in treatment in preclinical studies, intending to provide references for subsequent studies and clinical applications in the future.
Collapse
Affiliation(s)
- 欣越 王
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 志兴 牛
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 德茂 张
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 静 谢
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 学东 周
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Functionally diverse heteromeric traps for ligands of the transforming growth factor-β superfamily. Sci Rep 2021; 11:18341. [PMID: 34526551 PMCID: PMC8443706 DOI: 10.1038/s41598-021-97203-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/18/2021] [Indexed: 01/19/2023] Open
Abstract
Ligands of the transforming growth factor-β (TGF-β) superfamily are important targets for therapeutic intervention but present challenges because they signal combinatorially and exhibit overlapping activities in vivo. To obtain agents capable of sequestering multiple TGF-β superfamily ligands with novel selectivity, we generated soluble, heterodimeric ligand traps by pairing the extracellular domain (ECD) of the native activin receptor type IIB (ActRIIB) alternately with the ECDs of native type I receptors activin receptor-like kinase 4 (ALK4), ALK7, or ALK3. Systematic analysis of these heterodimeric constructs by surface plasmon resonance, and comparison with their homodimeric counterparts, revealed that each type I receptor partner confers a distinct ligand-binding profile to the heterodimeric construct. Additional characterization in cell-based reporter gene assays confirmed that the heterodimeric constructs possessed different profiles of signaling inhibition in vitro, which translated into altered patterns of pharmacological activity when constructs were administered systemically to wild-type mice. Our results detail a versatile platform for the modular recombination of naturally occurring receptor domains, giving rise to inhibitory ligand traps that could aid in defining the physiological roles of TGF-β ligand sets or be directed therapeutically to human diseases arising from dysregulated TGF-β superfamily signaling.
Collapse
|
24
|
Heubel B, Nohe A. The Role of BMP Signaling in Osteoclast Regulation. J Dev Biol 2021; 9:24. [PMID: 34203252 PMCID: PMC8293073 DOI: 10.3390/jdb9030024] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/02/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
The osteogenic effects of Bone Morphogenetic Proteins (BMPs) were delineated in 1965 when Urist et al. showed that BMPs could induce ectopic bone formation. In subsequent decades, the effects of BMPs on bone formation and maintenance were established. BMPs induce proliferation in osteoprogenitor cells and increase mineralization activity in osteoblasts. The role of BMPs in bone homeostasis and repair led to the approval of BMP2 by the Federal Drug Administration (FDA) for anterior lumbar interbody fusion (ALIF) to increase the bone formation in the treated area. However, the use of BMP2 for treatment of degenerative bone diseases such as osteoporosis is still uncertain as patients treated with BMP2 results in the stimulation of not only osteoblast mineralization, but also osteoclast absorption, leading to early bone graft subsidence. The increase in absorption activity is the result of direct stimulation of osteoclasts by BMP2 working synergistically with the RANK signaling pathway. The dual effect of BMPs on bone resorption and mineralization highlights the essential role of BMP-signaling in bone homeostasis, making it a putative therapeutic target for diseases like osteoporosis. Before the BMP pathway can be utilized in the treatment of osteoporosis a better understanding of how BMP-signaling regulates osteoclasts must be established.
Collapse
Affiliation(s)
- Brian Heubel
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Anja Nohe
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
25
|
Ledesma-Colunga MG, Weidner H, Vujic Spasic M, Hofbauer LC, Baschant U, Rauner M. Shaping the bone through iron and iron-related proteins. Semin Hematol 2021; 58:188-200. [PMID: 34389111 DOI: 10.1053/j.seminhematol.2021.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/18/2021] [Accepted: 06/08/2021] [Indexed: 01/04/2023]
Abstract
Well-controlled iron levels are indispensable for health. Iron deficiency is the most common cause of anemia, whereas iron overload, either hereditary or secondary due to disorders of ineffective erythropoiesis, causes widespread organ failure. Bone is particularly sensitive to fluctuations in systemic iron levels as both iron deficiency and overload are associated with low bone mineral density and fragility. Recent studies have shown that not only iron itself, but also iron-regulatory proteins that are mutated in hereditary hemochromatosis can control bone mass. This review will summarize the current knowledge on the effects of iron on bone homeostasis and bone cell activities, and on the role of proteins that regulate iron homeostasis, i.e. hemochromatosis proteins and proteins of the bone morphogenetic protein pathway, on bone remodeling. As disorders of iron homeostasis are closely linked to bone fragility, deeper insights into common regulatory mechanisms may provide new opportunities to concurrently treat disorders affecting iron homeostasis and bone.
Collapse
Affiliation(s)
- Maria G Ledesma-Colunga
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Heike Weidner
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Maja Vujic Spasic
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Lorenz C Hofbauer
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Divisions of Endocrinology and Molecular Bone Biology, Department of Medicine III & University Center for Healty Aging, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
26
|
Hu Y, Hao X, Liu C, Ren C, Wang S, Yan G, Meng Y, Mishina Y, Shi C, Sun H. Acvr1 deletion in osteoblasts impaired mandibular bone mass through compromised osteoblast differentiation and enhanced sRANKL-induced osteoclastogenesis. J Cell Physiol 2021; 236:4580-4591. [PMID: 33251612 PMCID: PMC8048423 DOI: 10.1002/jcp.30183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/30/2022]
Abstract
Bone morphogenetic protein (BMP) signaling is well known in bone homeostasis. However, the physiological effects of BMP signaling on mandibles are largely unknown, as the mandible has distinct functions and characteristics from other bones. In this study, we investigated the roles of BMP signaling in bone homeostasis of the mandibles by deleting BMP type I receptor Acvr1 in osteoblast lineage cells with Osterix-Cre. We found mandibular bone loss in conditional knockout mice at the ages of postnatal day 21 and 42 in an age-dependent manner. The decreased bone mass was related to compromised osteoblast differentiation together with enhanced osteoclastogenesis, which was secondary to the changes in osteoblasts in vivo. In vitro study revealed that deletion of Acvr1 in the mandibular bone marrow stromal cells (BMSCs) significantly compromised osteoblast differentiation. When wild type bone marrow macrophages were cocultured with BMSCs lacking Acvr1 both directly and indirectly, both proliferation and differentiation of osteoclasts were induced as evidenced by an increase of multinucleated cells, compared with cocultured with control BMSCs. Furthermore, we demonstrated that the increased osteoclastogenesis in vitro was at least partially due to the secretion of soluble receptor activator of nuclear factor-κB ligand (sRANKL), which is probably the reason for the mandibular bone loss in vivo. Overall, our results proposed that ACVR1 played essential roles in maintaining mandibular bone homeostasis through osteoblast differentiation and osteoblast-osteoclast communication via sRANKL.
Collapse
Affiliation(s)
- Yue Hu
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Xinqing Hao
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Cangwei Liu
- Department of Oral Pathology, School and Hospital of StomatologyChina Medical UniversityShenyangChina
| | - Chunxia Ren
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Shuangshuang Wang
- Department of Oral Pathology, School and Hospital of StomatologyChina Medical UniversityShenyangChina
| | - Guangxing Yan
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Yuan Meng
- Department of Oral Pathology, School and Hospital of StomatologyChina Medical UniversityShenyangChina
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of DentistryUniversity of MichiganAnn ArborMichiganUSA
| | - Ce Shi
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Hongchen Sun
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| |
Collapse
|
27
|
Song D, He G, Shi Y, Ni J, Long F. Functional interaction between Wnt and Bmp signaling in periosteal bone growth. Sci Rep 2021; 11:10782. [PMID: 34031510 PMCID: PMC8144582 DOI: 10.1038/s41598-021-90324-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/04/2021] [Indexed: 02/05/2023] Open
Abstract
Wnt and Bmp proteins are well known to regulate bone development and homeostasis. Although both signals are extensively studied, their potential interaction in vivo is less well understood. Previous studies have shown that deletion of Bmpr1a, a type I receptor for Bmp signaling, results in excessive trabecular bone formation while diminishing periosteal bone growth. Moreover, forced-expression of the Wnt antagonist Sost suppresses the overgrowth of trabecular bone caused by Bmpr1a deletion, thus implicating hyperactive Wnt signaling in the excessive trabecular bone formation. However, it remains uncertain whether Wnt and Bmp signaling interacts in regulating the periosteal bone growth. Here we show that multiple Wnt genes are markedly suppressed in the cortical bone without Bmpr1a. Importantly, overexpression of Wnt7b fully rescues periosteal bone growth in the Bmpr1a-deficient mice. Thus, pharmacological activation of Wnt signaling can restore normal bone size without intact Bmp signaling.
Collapse
Affiliation(s)
- Deye Song
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yu Shi
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiangdong Ni
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Fanxin Long
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA. .,Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA. .,Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Histone modifications centric-regulation in osteogenic differentiation. Cell Death Dis 2021; 7:91. [PMID: 33941771 PMCID: PMC8093204 DOI: 10.1038/s41420-021-00472-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/21/2021] [Accepted: 04/07/2021] [Indexed: 02/03/2023]
Abstract
Histone modification critically contributes to the epigenetic control of gene expression by changing the configuration of chromatin and modifying the access of transcription factors to gene promoters. Recently, we observed that histone acetylation and crotonylation mediated the expression of endocytosis-related genes and tumor-related immune checkpoint genes by regulating the enrichment of signal transducer and activator of transcription 3 on these gene promoters in Alzheimer's disease and tumorigenesis, suggesting that histone modification plays an important role in disease development. Furthermore, studies performed in the past decade revealed that histone modifications affect osteogenic differentiation by regulating the expression of osteogenic marker genes. In this review, we summarize and discuss the histone modification-centric regulation of osteogenic gene expression. This review improves the understanding of the role of histone modifications in osteogenic differentiation and describes its potential as a therapeutic target for osteogenic differentiation-related diseases.
Collapse
|
29
|
Alhasyimi AA, Suparwitri S, Christnawati C. Effect of Carbonate Apatite Hydrogel-Advanced Platelet-Rich Fibrin Injection on Osteoblastogenesis during Orthodontic Relapse in Rabbits. Eur J Dent 2020; 15:412-419. [PMID: 33368063 PMCID: PMC8382455 DOI: 10.1055/s-0040-1721234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective
This study aimed to determine the effect of carbonate apatite (CHA) hydrogel-aPRF on osteoblastogenesis during relapse in rabbits.
Materials and Methods
Forty-five rabbits were divided into three groups (
n
= 15): the control, CHA, and CHA-autologous platelet-rich fibrin (aPRF) groups. An open-coil spring was compressed between brackets to distalize the lower incisors of the rabbits by delivering a force of 50 cN for 1 week. The new position of the teeth was retained for 14 days, and CHA hydrogel-aPRF was injected every 7 days. The appliances were then debonded to allow relapse. On days 0, 3, 7, 14, and 21 after debonding, transforming growth factor (TGF)-β1 and bone morphogenetic protein (BMP)-2 expression was examined using immunohistochemistry staining and Runx-2 levels were analyzed by enzyme-linked immunosorbent assay. The data collected were analyzed using analysis of variance and a post hoc Tukey’s test (
p
< 0.05).
Results
Histomorphometric analysis revealed that TGF-β1 expression in the CHA-aPRF group is statistically higher than that in other groups on days 0, 3, and 7 after debonding (
p
< 0.05). BMP-2 expression in the CHA-aPRF group was also statistically higher than that in the other groups on days 3, 14, and 21 after debonding (
p
< 0.05). ELISA showed that Runx-2 levels are slightly higher in the CHA-aPRF group than in the other groups (
p
> 0.05).
Conclusion
Although injection of CHA-aPRF aids in osteoblastogenesis associated with enhancing TGF-β1 and BMP-2 expressions, it does not significantly upregulate Runx-2 levels.
Collapse
Affiliation(s)
- Ananto Ali Alhasyimi
- Department of Orthodontics, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Sri Suparwitri
- Department of Orthodontics, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | |
Collapse
|
30
|
Effect of immunosuppressants on a mouse model of osteogenesis imperfecta type V harboring a heterozygous Ifitm5 c.-14C > T mutation. Sci Rep 2020; 10:21197. [PMID: 33273604 PMCID: PMC7713238 DOI: 10.1038/s41598-020-78403-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/19/2020] [Indexed: 01/08/2023] Open
Abstract
Osteogenesis imperfecta (OI) type V is an autosomal dominant disorder caused by the c.-14C > T mutation in the interferon-induced transmembrane protein 5 gene (IFITM5), however, its onset mechanism remains unclear. In this study, heterozygous c.-14C > T mutant mice were developed to investigate the effect of immunosuppressants (FK506 and rapamycin) on OI type V. Among the mosaic mice generated by Crispr/Cas9-based technology, mice with less than 40% mosaic ratio of c.-14C > T mutation survived, whereas those with more than 48% mosaic ratio exhibited lethal skeletal abnormalities with one exception. All heterozygous mutants obtained by mating mosaic mice with wild-type mice exhibited a perinatal lethal phenotype due to severe skeletal abnormalities. Administration of FK506, a calcineurin inhibitor, in the heterozygous fetuses improved bone mineral content (BMC) of the neonates, although it did not save the neonates from the lethal effects of the mutation, whereas rapamycin, an mTOR inhibitor, reduced BMC, suggesting that mTOR signaling is involved in the bone mineralization of heterozygous mutants. These findings could clarify certain aspects of the onset mechanism of OI type V and enable development of therapeutics for this condition.
Collapse
|
31
|
Lademann F, Hofbauer LC, Rauner M. The Bone Morphogenetic Protein Pathway: The Osteoclastic Perspective. Front Cell Dev Biol 2020; 8:586031. [PMID: 33178699 PMCID: PMC7597383 DOI: 10.3389/fcell.2020.586031] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022] Open
Abstract
Bone health crucially relies on constant bone remodeling and bone regeneration, both tightly controlled processes requiring bone formation and bone resorption. Plenty of evidence identifies bone morphogenetic proteins (BMP) as major players in osteoblast differentiation and thus, bone formation. However, in recent past years, researchers also increasingly reported on the pivotal role of these multi-functional growth factors in osteoclast formation and activity. This review aims to summarize the current knowledge of BMP signaling within the osteoclast lineage, its role in bone resorption, and osteoblast-osteoclast coupling. Furthermore, subsequent clinical implications for recombinant BMP therapy will be discussed in view of recent preclinical and clinical studies.
Collapse
Affiliation(s)
- Franziska Lademann
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C. Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
32
|
Lademann F, Weidner H, Tsourdi E, Kumar R, Rijntjes E, Köhrle J, Hofbauer LC, Rauner M. Disruption of BMP Signaling Prevents Hyperthyroidism-Induced Bone Loss in Male Mice. J Bone Miner Res 2020; 35:2058-2069. [PMID: 32453466 DOI: 10.1002/jbmr.4092] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022]
Abstract
Thyroid hormones (TH) are key regulators of bone health, and TH excess in mice causes high bone turnover-mediated bone loss. However, the underlying molecular mechanisms of TH actions on bone remain poorly defined. Here, we tested the hypothesis whether TH mediate their effects via the pro-osteogenic bone morphogenetic protein (BMP) signaling pathway in vitro and in vivo. Primary murine osteoblasts treated with 3,3',5-triiodo-L-thyronine (T3 ) showed an enhanced differentiation potential, which was associated with activated canonical BMP/SMAD signaling reflected by SMAD1/5/8 phosphorylation. Blocking BMP signaling at the receptor (LDN193189) and ligand level (noggin, anti-BMP2/BMP4 neutralizing antibodies) inhibited T3 -induced osteogenic differentiation. In vivo, TH excess over 4 weeks in male C57BL/6JRj mice led to severe trabecular bone loss with a high bone turnover that was completely prevented by treatment with the BMP ligand scavenger ALK3-Fc. Thus, TH activate the canonical BMP pathway in osteoblasts to promote their differentiation and function. Importantly, this study indicates that blocking the BMP pathway may be an effective strategy to treat hyperthyroidism-induced bone loss. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Franziska Lademann
- Department of Medicine III, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany
| | - Heike Weidner
- Department of Medicine III, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany
| | - Elena Tsourdi
- Department of Medicine III, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ravi Kumar
- Acceleron Pharma, Inc, Cambridge, MA, USA
| | - Eddy Rijntjes
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Universitätsklinikum Dresden, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
33
|
BMP-induced Atoh8 attenuates osteoclastogenesis by suppressing Runx2 transcriptional activity and reducing the Rankl/Opg expression ratio in osteoblasts. Bone Res 2020; 8:32. [PMID: 32923015 PMCID: PMC7463266 DOI: 10.1038/s41413-020-00106-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 04/03/2020] [Accepted: 05/10/2020] [Indexed: 12/19/2022] Open
Abstract
Adult bone structural integrity is maintained by remodeling via the coupling of osteoclastic bone resorption and osteoblastic bone formation. Osteocytes or osteoblasts express receptor activator of nuclear factor κ-B ligand (Rankl) or osteoprotegerin (Opg) to promote or inhibit osteoclastogenesis, respectively. Bone morphogenetic protein (BMP) is a potent bone inducer, but its major role in adult bone is to induce osteocytes to upregulate sclerostin (Sost) and increase the Rankl/Opg expression ratio, resulting in promotion of osteoclastogenesis. However, the precise effect of BMP-target gene(s) in osteoblasts on the Rankl/Opg expression ratio remains unclear. In the present study, we identified atonal homolog 8 (Atoh8), which is directly upregulated by the BMP-Smad1 axis in osteoblasts. In vivo, Atoh8 was detected in osteoblasts but not osteocytes in adult mice. Although global Atoh8-knockout mice showed only a mild phenotype in the neonate skeleton, the bone volume was decreased and osteoclasts were increased in the adult phase. Atoh8-null marrow stroma cells were more potent than wild-type cells in inducing osteoclastogenesis in marrow cells. Atoh8 loss in osteoblasts increased Runx2 expression and the Rankl/Opg expression ratio, while Runx2 knockdown normalized the Rankl/Opg expression ratio. Moreover, Atoh8 formed a protein complex with Runx2 to inhibit Runx2 transcriptional activity and decrease the Rankl/Opg expression ratio. These results suggest that bone remodeling is regulated elaborately by BMP signaling; while BMP primarily promotes bone resorption, it simultaneously induces Atoh8 to inhibit Runx2 and reduce the Rankl/Opg expression ratio in osteoblasts, suppressing osteoclastogenesis and preventing excessive BMP-mediated bone resorption.
Collapse
|
34
|
Kamiya N, Atsawasuwan P, Joiner DM, Waldorff EI, Goldstein S, Yamauchi M, Mishina Y. Controversy of physiological vs. pharmacological effects of BMP signaling: Constitutive activation of BMP type IA receptor-dependent signaling in osteoblast lineage enhances bone formation and resorption, not affecting net bone mass. Bone 2020; 138:115513. [PMID: 32603910 PMCID: PMC7423725 DOI: 10.1016/j.bone.2020.115513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022]
Abstract
Bone morphogenetic proteins (BMPs) were first described over 50 years ago as potent inducers of ectopic bone formation when administrated subcutaneously. Preclinical studies have extensively examined the osteoinductive properties of BMPs in vitro and new bone formation in vivo. BMPs (BMP-2, BMP-7) have been used in orthopedics over 15 years. While osteogenic function of BMPs has been widely accepted, our previous studies demonstrated that loss-of-function of BMP receptor type IA (BMPR1A), a potent receptor for BMP-2, increased net bone mass by significantly inhibiting bone resorption in mice, indicating a positive role of BMP signaling in bone resorption. The physiological role of BMPs (i.e. osteogenic vs. osteoclastogenic) is still largely unknown. The purpose of this study was to investigate the physiological role of BMP signaling in endogenous long bones during adult stages. For this purpose, we conditionally and constitutively activated the Smad-dependent canonical BMP signaling thorough BMPR1A in osteoblast lineage cells using the mutant mice (Col1CreER™:caBmpr1a). Because trabecular bones were largely increased in the loss-of-function mouse study for BMPR1A, we hypothesized that the augmented BMP signaling would affect endogenous trabecular bones. In the mutant bones, the Smad phosphorylation was enhanced within physiological level three-fold while the resulting gross morphology, bodyweights, bone mass/shape/length, serum calcium/phosphorus levels, collagen cross-link patterns, and healing capability were all unchanged. Interestingly, we found; 1) increased expressions of both bone formation and resorption markers in femoral bones, 2) increased osteoblast and osteoclast numbers together with dynamic bone formation parameters by trabecular bone histomorphometry, 3) modest bone architectural phenotype with reduced bone quality (i.e. reduced trabecular bone connectivity, larger diametric size but reduced cortical bone thickness, and reduced bone mechanical strength), and 4) increased expression of SOST, a downstream target of the Smad-dependent BMPR1A signaling, in the mutant bones. This study is clinically insightful because gain-of-function of BMP signaling within a physiological window does not increase bone mass while it alters molecular and cellular aspects of osteoblast and osteoclast functions as predicted. These findings help explain the high-doses of BMPs (i.e. pharmacological level) in clinical settings required to substantially induce a bone formation, concurrent with potential unexpected side effects (i.e. bone resorption, inflammation) presumably due to a broader population of cell-types exposed to the high-dose BMPs rather than osteoblastic lineage cells.
Collapse
Affiliation(s)
- Nobuhiro Kamiya
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA; Faculty of Budo and Sport Studies, Tenri University, Nara 6320071, Japan; Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA.
| | - Phimon Atsawasuwan
- School of Dentistry, University of North Carolina, Chapel Hill, NC 27599, USA; College of Dentistry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Danese M Joiner
- Department of Orthopaedic Surgery, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Erik I Waldorff
- Department of Orthopaedic Surgery, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Steve Goldstein
- Department of Orthopaedic Surgery, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Mitsuo Yamauchi
- School of Dentistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA.
| |
Collapse
|
35
|
Zhang H, Zhang Y, Terajima M, Romanowicz G, Liu Y, Omi M, Bigelow E, Joiner DM, Waldorff EI, Zhu P, Raghavan M, Lynch M, Kamiya N, Zhang R, Jepsen KJ, Goldstein S, Morris MD, Yamauchi M, Kohn DH, Mishina Y. Loss of BMP signaling mediated by BMPR1A in osteoblasts leads to differential bone phenotypes in mice depending on anatomical location of the bones. Bone 2020; 137:115402. [PMID: 32360900 PMCID: PMC7354232 DOI: 10.1016/j.bone.2020.115402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022]
Abstract
Bone morphogenetic protein (BMP) signaling in osteoblasts plays critical roles in skeletal development and bone homeostasis. Our previous studies showed loss of function of BMPR1A, one of the type 1 receptors for BMPs, in osteoblasts results in increased trabecular bone mass in long bones due to an imbalance between bone formation and bone resorption. Decreased bone resorption was associated with an increased mature-to-immature collagen cross-link ratio and mineral-matrix ratios in the trabecular compartments, and increased tissue-level biomechanical properties. Here, we investigated the bone mass, bone composition and biomechanical properties of ribs and spines in the same genetically altered mouse line to compare outcomes by loss of BMPR1A functions in bones from different anatomic sites and developmental origins. Bone mass was significantly increased in both cortical and trabecular compartments of ribs with minimal to modest changes in compositions. While tissue-levels of biomechanical properties were not changed between control and mutant animals, whole bone levels of biomechanical properties were significantly increased in association with increased bone mass in the mutant ribs. For spines, mutant bones showed increased bone mass in both cortical and trabecular compartments with an increase of mineral content. These results emphasize the differential role of BMP signaling in osteoblasts in bones depending on their anatomical locations, functional loading requirements and developmental origin.
Collapse
Affiliation(s)
- Honghao Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Yanshuai Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Masahiko Terajima
- School of Dentistry, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - Genevieve Romanowicz
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Yangjia Liu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA; School of Life Sciences, Tsinghua University, Beijing, China
| | - Maiko Omi
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Erin Bigelow
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Danese M Joiner
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Erik I Waldorff
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Peizhi Zhu
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Mekhala Raghavan
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Michelle Lynch
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Nobuhiro Kamiya
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA; Tenri University, Nara, Japan
| | - Rongqing Zhang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Karl J Jepsen
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Steve Goldstein
- Department of Orthopaedic Surgery, Michigan Medicine, University of Michigan, MI, USA
| | - Michael D Morris
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Mitsuo Yamauchi
- School of Dentistry, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - David H Kohn
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA.
| |
Collapse
|
36
|
Qian Z, Zhang Y, Kang X, Li H, Zhang Y, Jin X, Gao X, Xu M, Ma Z, Zhao L, Zhang Z, Sun H, Wu S. Postnatal Conditional Deletion of Bmal1 in Osteoblasts Enhances Trabecular Bone Formation Via Increased BMP2 Signals. J Bone Miner Res 2020; 35:1481-1493. [PMID: 32212389 DOI: 10.1002/jbmr.4017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 03/09/2020] [Accepted: 03/20/2020] [Indexed: 01/09/2023]
Abstract
A large number of studies in recent years indicated the involvement of peripheral circadian clock in varied pathologies. However, evidence regarding how peripheral clocks regulate bone metabolism is still very limited. The present study aimed to investigate the direct role of Bmal1 (the key activator of peripheral circadian clock system) in vivo during bone developmental and remodeling stages using inducible osteoblast-specific Bmal1 knockout mice. Unexpectedly, the removal of Bmal1 in osteoblasts caused multiple abnormalities of bone metabolism, including a progressive increase in trabecular bone mass in as early as 8 weeks, manifested by an 82.3% increase in bone mineral density and 2.8-fold increase in bone volume per tissue volume. As mice age, an increase in trabecular bone mass persists while cortical bone mass decreases by about 33.7%, concomitant with kyphoscoliosis and malformed intervertebral disk. The increased trabecular bone mass is attributed to increased osteoblast number and osteoblast activity coupled with decreased osteoclastogenesis. Remarkably, the ablation of Bmal1 in osteoblasts promoted the expression level of Bmp2 and phosphorylation of SMAD1, whereas the attenuation of BMP2/SMAD1 signaling partially alleviated the effects of Bmal1 deficiency on osteoblast differentiation and activity. The results revealed that Bmal1 was a transcriptional silencer of Bmp2 by targeting the Bmp2 promoter. The peripheral clock gene Bmal1 in osteoblasts was crucial to coordinate differential effects on trabecular and cortical bones through regulating BMP2/SMAD1 during bone development, thus providing novel insights into a key role of osteoblast Bmal1 in homeostasis and integrity of adult bones. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Zhuang Qian
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Ying Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xiaomin Kang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Huixia Li
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xinxin Jin
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xin Gao
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Mao Xu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Zhengmin Ma
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Liting Zhao
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Zhuanmin Zhang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Hongzhi Sun
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Shufang Wu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
37
|
Jing Y, Wang Z, Li H, Ma C, Feng J. Chondrogenesis Defines Future Skeletal Patterns Via Cell Transdifferentiation from Chondrocytes to Bone Cells. Curr Osteoporos Rep 2020; 18:199-209. [PMID: 32219639 PMCID: PMC7717675 DOI: 10.1007/s11914-020-00586-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to obtain a better understanding of how chondrogenesis defines skeletal development via cell transdifferentiation from chondrocytes to bone cells. RECENT FINDINGS A breakthrough in cell lineage tracing allows bone biologists to trace the cell fate and demonstrate that hypertrophic chondrocytes can directly transdifferentiate into bone cells during endochondral bone formation. However, there is a knowledge gap for the biological significance of this lineage extension and the mechanisms controlling this process. This review first introduces the history of the debate on the cell fate of chondrocytes in endochondral bone formation; then summarizes key findings obtained in recent years, which strongly support a new theory: the direct cell transdifferentiation from chondrocytes to bone cells precisely connects chondrogenesis (for providing a template of the future skeleton, classified as phase I) and osteogenesis (for finishing skeletal construction, or phase II) in a continuous lineage-linked process of endochondral bone formation and limb elongation; and finally outlines nutrition factors and molecules that regulate the cell transdifferentiation process during the relay from chondrogenesis to osteogenesis.
Collapse
Affiliation(s)
- Yan Jing
- Department of Orthodontics, Texas A&M University College of Dentistry, 3302 Gaston ave, Dallas, TX, 75246, USA.
| | - Zheng Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Hui Li
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
- State Key Laboratory of Oral Diseases, Department of Traumatic and Plastic Surgery, , West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chi Ma
- Department of Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Jian Feng
- Department of Orthodontics, Texas A&M University College of Dentistry, 3302 Gaston ave, Dallas, TX, 75246, USA.
| |
Collapse
|
38
|
Ihle CL, Straign DM, Provera MD, Novitskiy SV, Owens P. Loss of Myeloid BMPR1a Alters Differentiation and Reduces Mouse Prostate Cancer Growth. Front Oncol 2020; 10:357. [PMID: 32318332 PMCID: PMC7154049 DOI: 10.3389/fonc.2020.00357] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/28/2020] [Indexed: 12/11/2022] Open
Abstract
The Bone Morphogenetic Protein (BMP) pathway is a member of the TGFβ signaling family and has complex roles in cancer. BMP signaling is rarely mutated and can be frequently overexpressed in many human cancers. The dichotomous role of BMPs as both tumor promoters and suppressors appears to be largely context based in both the cancer cell and the surrounding microenvironment. Myeloid cells including macrophages and neutrophils have been shown to be tumor promoting when stimulated from BMPs. We found that conditional deletion of BMPR1a in myeloid cells (LysMCre) restricts tumor progression in a syngeneic mouse prostate cancer model. Specific changes occurred in myeloid cells both in tumor bearing mice and tumor naïve mice throughout multiple tissues. We profiled myeloid subsets in the bone marrow, spleen and primary tumor and found myeloid BMPR1a loss altered the differentiation and lineage capability of distinct populations by histologic, flow cytometry and high dimensional mass cytometry analysis. We further confirmed the requirement for BMP signaling with pharmacologic inhibition of THP-1 and Raw264.7 activated into M2 macrophages with the BMP inhibitor DMH1. M2 polarized primary bone marrow derived cells from LysMCre BMPR1a knockout mice indicated a distinct requirement for BMP signaling in myeloid cells during M2 activation. These results indicate a unique necessity for BMP signaling in myeloid cells during tumor progression.
Collapse
Affiliation(s)
- Claire L. Ihle
- Cancer Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Desiree M. Straign
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Meredith D. Provera
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sergey V. Novitskiy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Philip Owens
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Veterans Affairs, Research Service, Eastern Colorado Health Care System, Aurora, CO, United States
| |
Collapse
|
39
|
Wang S, Li J, Sun H, Sha L, Guo Y, Gu G, Mao J, Nie X, Zhai Y, Yu D, Zhai J, Li H, Shan X, Dai C, Wu X, He X, Xin L, Liu J, Heng K, Geng Q. Treatment with soluble bone morphogenetic protein type 1A receptor fusion protein alleviates irradiation-induced bone loss in mice through increased bone formation and reduced bone resorption. Am J Transl Res 2020; 12:743-757. [PMID: 32269709 PMCID: PMC7137047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 12/23/2019] [Indexed: 06/11/2023]
Abstract
An increased fracture risk is often observed in cancer patients undergoing radiotherapy (RT), particularly at sites within the field of radiation. Therefore, the development of appropriate therapeutic options to prevent RT-induced bone loss is urgently needed. A soluble form of the BMP receptor type 1A fusion protein (mBMPR1A-mFc) serves as an antagonist to endogenous BMPR1A. Previous studies have shown that mBMPR1A-mFc treatment increases bone mass in both ovary-intact and ovariectomized via promoting osteoblastic bone formation and inhibiting osteoclastic bone resorption. The present study was designed to investigate whether mBMPR1A-mFc administration prevents radiation-induced bone deterioration in mice. We constructed an animal model of radiation-induced osteoporosis by exposure to a 2-Gy dose of X-rays. Micro-CT, histomorphometric, bone-turnover, and mechanical analyses showed that mBMPR1A-mFc administration prevented trabecular microarchitecture deterioration after RT because of a marked increase in bone formation and a decrease in bone resorption. Mechanistic studies indicated that mBMPR1A-mFc administration promoted osteoblastogenesis by activating Wnt/Lrp5/β-catenin signaling while decreasing osteoclastogenesis by inhibiting the RANKL/RANK/OPG pathway. Our novel findings provide solid evidence for the application of mBMPR1A-mFc as a therapeutic treatment for radiation-induced osteoporosis.
Collapse
Affiliation(s)
- Shen Wang
- Department of Acupuncture, Guangxi Medical UniversityNanning 530000, China
| | - Jie Li
- Department of Orthopedics, Xuzhou Central Hospital, Southeast UniversityXuzhou 221006, China
| | - Huabei Sun
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Liangwei Sha
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Yilong Guo
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Guanqiu Gu
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Jiling Mao
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Xinfa Nie
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Ying Zhai
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Dehong Yu
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Juan Zhai
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Hongnian Li
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Xin Shan
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Chengbai Dai
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Xiangzhi Wu
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Xiaobo He
- Department of Orthopedics, Yangzhou Jiangdu TCM HospitalYangzhou 225200, China
| | - Li Xin
- Department of Osteoporosis, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou 221004, China
| | - Jun Liu
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical UniversityNanjing 210011, China
| | - Ke Heng
- Department of Orthopedics, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical UniversityChangzhou 213003, China
| | - Qinghe Geng
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| |
Collapse
|
40
|
Chen G, Xu H, Yao Y, Xu T, Yuan M, Zhang X, Lv Z, Wu M. BMP Signaling in the Development and Regeneration of Cranium Bones and Maintenance of Calvarial Stem Cells. Front Cell Dev Biol 2020; 8:135. [PMID: 32211409 PMCID: PMC7075941 DOI: 10.3389/fcell.2020.00135] [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: 12/27/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
The bone morphogenetic protein (BMP) signaling pathway is highly conserved across many species, and its importance for the patterning of the skeletal system has been demonstrated. A disrupted BMP signaling pathway results in severe skeletal defects. Murine calvaria has been identified to have dual-tissue lineages, namely, the cranial neural-crest cells and the paraxial mesoderm. Modulations of the BMP signaling pathway have been demonstrated to be significant in determining calvarial osteogenic potentials and ossification in vitro and in vivo. More importantly, the BMP signaling pathway plays a role in the maintenance of the homeostasis of the calvarial stem cells, indicating a potential clinic significance in calvarial bone and in expediting regeneration. Following the inherent evidence of BMP signaling in craniofacial biology, we summarize recent discoveries relating to BMP signaling in the development of calvarial structures, functions of the suture stem cells and their niche and regeneration. This review will not only provide a better understanding of BMP signaling in cranial biology, but also exhibit the molecular targets of BMP signaling that possess clinical potential for tissue regeneration.
Collapse
Affiliation(s)
- Guiqian Chen
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Haodong Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yifeng Yao
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Tingting Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Mengting Yuan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xingen Zhang
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Zhengbing Lv
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Mengrui Wu
- Institute of Genetics, Life Science College, Zhejiang University, Hangzhou, China
| |
Collapse
|
41
|
Maridas DE, Feigenson M, Renthal NE, Chim SM, Gamer LW, Rosen V. Bone morphogenetic proteins. PRINCIPLES OF BONE BIOLOGY 2020:1189-1197. [DOI: 10.1016/b978-0-12-814841-9.00048-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Westhrin M, Holien T, Zahoor M, Moen SH, Buene G, Størdal B, Hella H, Yuan H, de Bruijn JD, Martens A, Groen RW, Bosch F, Smith U, Sponaas AM, Sundan A, Standal T. Bone Morphogenetic Protein 4 Gene Therapy in Mice Inhibits Myeloma Tumor Growth, But Has a Negative Impact on Bone. JBMR Plus 2019; 4:e10247. [PMID: 31956851 PMCID: PMC6957984 DOI: 10.1002/jbm4.10247] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 02/03/2023] Open
Abstract
Multiple myeloma is characterized by accumulation of malignant plasma cells in the bone marrow. Most patients suffer from an osteolytic bone disease, caused by increased bone degradation and reduced bone formation. Bone morphogenetic protein 4 (BMP4) is important for both pre‐ and postnatal bone formation and induces growth arrest and apoptosis of myeloma cells. BMP4‐treatment of myeloma patients could have the potential to reduce tumor growth and restore bone formation. We therefore explored BMP4 gene therapy in a human‐mouse model of multiple myeloma where humanized bone scaffolds were implanted subcutaneously in RAG2−/− γC−/−mice. Mice were treated with adeno‐associated virus serotype 8 BMP4 vectors (AAV8‐BMP4) to express BMP4 in the liver. When mature BMP4 was detectable in the circulation, myeloma cells were injected into the scaffolds and tumor growth was examined by weekly imaging. Strikingly, the tumor burden was reduced in AAV8‐BMP4 mice compared with the AAV8‐CTRL mice, suggesting that increased circulating BMP4 reduced tumor growth. BMP4‐treatment also prevented bone loss in the scaffolds, most likely due to reduced tumor load. To delineate the effects of BMP4 overexpression on bone per se, without direct influence from cancer cells, we examined the unaffected, non‐myeloma femurs by μCT. Surprisingly, the AAV8‐BMP4 mice had significantly reduced trabecular bone volume, trabecular numbers, as well as significantly increased trabecular separation compared with the AAV8‐CTRL mice. There was no difference in cortical bone parameters between the two groups. Taken together, BMP4 gene therapy inhibited myeloma tumor growth, but also reduced the amount of trabecular bone in mice. Our data suggest that care should be taken when considering using BMP4 as a therapeutic agent. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marita Westhrin
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway.,Centre of Molecular Inflammation Research (CEMIR) Norwegian University of Science and Technology Trondheim Norway
| | - Toril Holien
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway.,Department of Hematology St. Olavs Hospital Trondheim Norway
| | - Muhammad Zahoor
- Centre of Molecular Inflammation Research (CEMIR) Norwegian University of Science and Technology Trondheim Norway
| | - Siv Helen Moen
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway.,Centre of Molecular Inflammation Research (CEMIR) Norwegian University of Science and Technology Trondheim Norway
| | - Glenn Buene
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway.,Centre of Molecular Inflammation Research (CEMIR) Norwegian University of Science and Technology Trondheim Norway
| | - Berit Størdal
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway
| | - Hanne Hella
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway
| | - Huipin Yuan
- Kuros Biosciences BV Bilthoven The Netherlands
| | - Joost D de Bruijn
- Kuros Biosciences BV Bilthoven The Netherlands.,The School of Engineering and Materials Science Queen Mary University of London London UK
| | - Anton Martens
- Department of Hematology Cancer Center Amsterdam, VU University Medical Center Amsterdam The Netherlands
| | - Richard Wj Groen
- Department of Hematology Cancer Center Amsterdam, VU University Medical Center Amsterdam The Netherlands
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology School of Veterinary Medicine, Universitat Autònoma de Barcelona Barcelona Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) Madrid Spain
| | - Ulf Smith
- Department of Molecular and Clinical Medicine Sahlgrenska University Hospital Gothenburg Sweden
| | - Anne-Marit Sponaas
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway
| | - Anders Sundan
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway.,Centre of Molecular Inflammation Research (CEMIR) Norwegian University of Science and Technology Trondheim Norway
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Faculty of Medicine Norwegian University of Science and Technology (NTNU) Trondheim Norway.,Centre of Molecular Inflammation Research (CEMIR) Norwegian University of Science and Technology Trondheim Norway.,Department of Hematology St. Olavs Hospital Trondheim Norway
| |
Collapse
|
43
|
Omi M, Kaartinen V, Mishina Y. Activin A receptor type 1-mediated BMP signaling regulates RANKL-induced osteoclastogenesis via canonical SMAD-signaling pathway. J Biol Chem 2019; 294:17818-17836. [PMID: 31619522 DOI: 10.1074/jbc.ra119.009521] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are important mediators of osteoclast differentiation. Although accumulating evidence has implicated BMPs in osteoblastogenesis, the mechanisms by which BMPs regulate osteoclastogenesis remain unclear. Activin A receptor type 1 (ACVR1) is a BMP type 1 receptor essential for skeletal development. Here, we observed that BMP-7, which preferentially binds to ACVR1, promotes osteoclast differentiation, suggesting ACVR1 is involved in osteoclastogenesis. To investigate this further, we isolated osteoclasts from either Acvr1-floxed mice or mice with constitutively-activated Acvr1 (caAcvr1) carrying tamoxifen-inducible Cre driven by a ubiquitin promotor and induced Cre activity in culture. Osteoclasts from the Acvr1-floxed mice had reduced osteoclast numbers and demineralization activity, whereas those from the caAcvr1-mutant mice formed large osteoclasts and demineralized pits, suggesting that BMP signaling through ACVR1 regulates osteoclast fusion and activity. It is reported that BMP-2 binds to BMPR1A, another BMP type 1 receptor, whereas BMP-7 binds to ACVR1 to activate SMAD1/5/9 signaling. Here, Bmpr1a-disrupted osteoclasts displayed reduced phospho-SMAD1/5/9 (pSMAD1/5/9) levels when induced by BMP-2, whereas no impacts on pSMAD1/5/9 were observed when induced by BMP-7. In contract, Acvr1-disrupted osteoclasts displayed reduced pSMAD1/5/9 levels when induced either by BMP-2 or BMP-7, suggesting that ACVR1 is the major receptor for transducing BMP-7 signals in osteoclasts. Indeed, LDN-193189 and LDN-212854, which specifically block SMAD1/5/9 phosphorylation, inhibited osteoclastogenesis of caAcvr1-mutant cells. Moreover, increased BMP signaling promoted nuclear translocation of nuclear factor-activated T-cells 1 (NFATc1), which was inhibited by LDN treatments. Taken together, ACVR1-mediated BMP-SMAD signaling activates NFATc1, a regulatory protein crucial for receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis.
Collapse
Affiliation(s)
- Maiko Omi
- Department of Biologic and Materials Sciences and Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences and Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Yuji Mishina
- Department of Biologic and Materials Sciences and Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
44
|
Gooding S, Olechnowicz SWZ, Morris EV, Armitage AE, Arezes J, Frost J, Repapi E, Edwards JR, Ashley N, Waugh C, Gray N, Martinez-Hackert E, Lim PJ, Pasricha SR, Knowles H, Mead AJ, Ramasamy K, Drakesmith H, Edwards CM. Transcriptomic profiling of the myeloma bone-lining niche reveals BMP signalling inhibition to improve bone disease. Nat Commun 2019; 10:4533. [PMID: 31586071 PMCID: PMC6778199 DOI: 10.1038/s41467-019-12296-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 08/30/2019] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma is an incurable, bone marrow-dwelling malignancy that disrupts bone homeostasis causing skeletal damage and pain. Mechanisms underlying myeloma-induced bone destruction are poorly understood and current therapies do not restore lost bone mass. Using transcriptomic profiling of isolated bone lining cell subtypes from a murine myeloma model, we find that bone morphogenetic protein (BMP) signalling is upregulated in stromal progenitor cells. BMP signalling has not previously been reported to be dysregulated in myeloma bone disease. Inhibition of BMP signalling in vivo using either a small molecule BMP receptor antagonist or a solubilized BMPR1a-FC receptor ligand trap prevents trabecular and cortical bone volume loss caused by myeloma, without increasing tumour burden. BMP inhibition directly reduces osteoclastogenesis, increases osteoblasts and bone formation, and suppresses bone marrow sclerostin levels. In summary we describe a novel role for the BMP pathway in myeloma-induced bone disease that can be therapeutically targeted.
Collapse
Affiliation(s)
- Sarah Gooding
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre Blood Theme, University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
| | - Sam W Z Olechnowicz
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
- Nuffield Dept. of Surgical Sciences, University of Oxford, Oxford, UK
| | - Emma V Morris
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
- Nuffield Dept. of Surgical Sciences, University of Oxford, Oxford, UK
| | - Andrew E Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Joao Arezes
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Joe Frost
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Emmanouela Repapi
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - James R Edwards
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
- Nuffield Dept. of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Neil Ashley
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Craig Waugh
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicola Gray
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Erik Martinez-Hackert
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Pei Jin Lim
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Sant-Rayn Pasricha
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Helen Knowles
- Nuffield Dept. of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Adam J Mead
- Oxford University Hospitals NHS Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre Blood Theme, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Karthik Ramasamy
- Oxford University Hospitals NHS Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre Blood Theme, University of Oxford, Oxford, UK
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre Blood Theme, University of Oxford, Oxford, UK.
| | - Claire M Edwards
- NIHR Oxford Biomedical Research Centre Blood Theme, University of Oxford, Oxford, UK.
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, UK.
- Nuffield Dept. of Surgical Sciences, University of Oxford, Oxford, UK.
- Nuffield Dept. of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
45
|
Geng Q, Heng K, Li J, Wang S, Sun H, Sha L, Guo Y, Nie X, Wang Q, Dai L, Zhu X, Kang J, Shao L, Zhai J, Miao S, Lin Q, Guo K, Wang J. A soluble bone morphogenetic protein type 1A receptor fusion protein treatment prevents glucocorticoid-Induced bone loss in mice. Am J Transl Res 2019; 11:4232-4247. [PMID: 31396331 PMCID: PMC6684880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is a frequent complication of systemic glucocorticoid (GC) therapy, is the most common form of secondary osteoporosis, and is associated with skeletal fragility and increased fracture risk. A soluble form of BMP receptor type 1A fusion protein (mBMPR1A-mFc) acts as an antagonist to endogenous BMPR1A and could increase bone mass in both ovariectomized and ovary-intact mice, but its effects in GIOP mice remained unclear. The aim of this study was to evaluate the effects of mBMPR1A-mFc on the skeleton in experimental models of GIOP. mBMPR1A-mFc treatment could increase the bone mineral density (BMD), trabecular bone volume, thickness, and number, and cortical thickness, and reduce the structure model index and trabecular separation in GIOP mice. mBMPR1A-mFc treatment could also prevent bone loss and enhance biomechanical strength in GIOP mice by promoting osteoblastic bone formation and inhibiting osteoclastic bone resorption. Mechanistic studies revealed that mBMPR1A-mFc treatment increased murine osteoblastogenesis by activating the Wnt/β-catenin signaling pathway while decreasing osteoclastogenesis by inhibiting the RANK/RANKL/osteoprotegerin (OPG) signaling pathway. These findings demonstrate that mBMPR1A-mFc treatment in GIOP mice improves bone mass, microarchitecture, and strength by enhancing osteoblastic bone formation and inhibiting osteoclastic bone resorption in GIOP mice and offers a promising novel alternative for the treatment of GIOP.
Collapse
Affiliation(s)
- Qinghe Geng
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Ke Heng
- Department of Orthopedics, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical UniversityChangzhou 213003, China
| | - Jie Li
- Department of Orthopedics, Xuzhou Central Hospital, Southeast UniversityXuzhou 221006, China
| | - Shen Wang
- Department of Acupuncture, Guangxi Medical UniversityNanning 530000, China
| | - Huabei Sun
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Liangwei Sha
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Yilong Guo
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Xinfa Nie
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Qingjun Wang
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Lei Dai
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Xianzhong Zhu
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Jiujie Kang
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Liwu Shao
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Juan Zhai
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| | - Sheng Miao
- Department of Orthopedics, The First Hospital of Suqian, Jiangsu Province Hospital, Nanjing Medical UniversitySuqian 223899, China
| | - Qiang Lin
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou 221004, China
| | - Kaijin Guo
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical UniversityXuzhou 221004, China
| | - Jin Wang
- Lab of Bone and Mineral Research, The Affiliated Pizhou Hospital of Xuzhou Medical UniversityXuzhou 221300, China
| |
Collapse
|
46
|
Moghaddam T, Neshati Z. Role of microRNAs in osteogenesis of stem cells. J Cell Biochem 2019; 120:14136-14155. [PMID: 31069839 DOI: 10.1002/jcb.28689] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/21/2022]
Abstract
Osteogenic differentiation is a controlled developmental process in which external and internal factors including cytokines, growth factors, transcription factors (TFs), signaling pathways and microRNAs (miRNAs) play important roles. Various stimulatory and inhibitory TFs contribute to osteogenic differentiation and are responsible for bone development. In addition, cross-talk between several complex signaling pathways regulates the osteogenic differentiation of some stem cells. Although much is known about regulatory genes and signaling pathways in osteogenesis, the role of miRNAs in osteogenic differentiation still needs to be explored. miRNAs are small, approximately 22 nucleotides, single-stranded nonprotein coding RNAs which are abundant in many mammalian cell types. They paly significant regulated roles in various biological processes and serve as promising biomarkers for disease states. Recently, emerging evidence have shown that miRNAs are the key regulators of osteogenesis of stem cells. They may endogenously regulate osteogenic differentiation of stem cells through direct targeting of positive or negative directors of osteogenesis and depending on the target result in the promotion or inhibition of osteogenic differentiation. This review aims to provide a general overview of miRNAs participating in osteogenic differentiation of stem cells and explain their regulatory effect based on the genes targeted with these miRNAs.
Collapse
Affiliation(s)
- Tayebe Moghaddam
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Zeinab Neshati
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
47
|
Bone morphogenetic proteins: Their role in regulating osteoclast differentiation. Bone Rep 2019; 10:100207. [PMID: 31193008 PMCID: PMC6513777 DOI: 10.1016/j.bonr.2019.100207] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 04/09/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023] Open
Abstract
The ability to create recombinant bone morphogenetic proteins (BMPs) in recent years has led to their rise as a common clinical adjuvant. Their application varies, from spinal fixation to repairing palatal clefts, to coating implants for osseointegration. In recent years questions have been raised as to the efficacy of BMPs in several of these procedures. These questions are due to the unwanted side effect of BMPs on other cell types, such as osteoclasts which can resorb bone at the graft/implant site. However, most BMP research focuses on the anabolic osteoinductive effects of BMPs on osteoblasts rather than its counterpart- stimulation of the osteoclasts, which are cells responsible for resorbing bone. In this review, we discuss the data available from multiple in-vitro and in-vivo BMP-related knockout models to elucidate the different functions BMPs have on osteoclast differentiation and activity. BMPs can act directly on osteoclasts to regulate differentiation and activity. Osteoclasts express multiple BMP signaling components. BMPs signal through both SMAD independent and dependent mechanisms in osteoclasts. SMAD dependent BMP signaling regulates osteoclast-osteoblast coupling factors.
Collapse
|
48
|
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor-β family of ligands. BMPs exhibit widespread utility and pleiotropic, context-dependent effects, and the strength and duration of BMP pathway signaling is tightly regulated at numerous levels via mechanisms operating both inside and outside the cell. Defects in the BMP pathway or its regulation underlie multiple human diseases of different organ systems. Yet much remains to be discovered about the BMP pathway in its original context, i.e., the skeleton. In this review, we provide a comprehensive overview of the intricacies of the BMP pathway and its inhibitors in bone development, homeostasis, and disease. We frame the content of the review around major unanswered questions for which incomplete evidence is available. First, we consider the gene regulatory network downstream of BMP signaling in osteoblastogenesis. Next, we examine why some BMP ligands are more osteogenic than others and what factors limit BMP signaling during osteoblastogenesis. Then we consider whether specific BMP pathway components are required for normal skeletal development, and if the pathway exerts endogenous effects in the aging skeleton. Finally, we propose two major areas of need of future study by the field: greater resolution of the gene regulatory network downstream of BMP signaling in the skeleton, and an expanded repertoire of reagents to reliably and specifically inhibit individual BMP pathway components.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| | - Vicki Rosen
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| |
Collapse
|
49
|
Zhang X, Shi C, Zhao H, Zhou Y, Hu Y, Yan G, Liu C, Li D, Hao X, Mishina Y, Liu Q, Sun H. Distinctive role of ACVR1 in dentin formation: requirement for dentin thickness in molars and prevention of osteodentin formation in incisors of mice. J Mol Histol 2018; 50:43-61. [PMID: 30519900 DOI: 10.1007/s10735-018-9806-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 11/29/2018] [Indexed: 11/24/2022]
Abstract
Dentin is a major component of teeth that protects dental pulp and maintains tooth health. Bone morphogenetic protein (BMP) signaling is required for the formation of dentin. Mice lacking a BMP type I receptor, activin A receptor type 1 (ACVR1), in the neural crest display a deformed mandible. Acvr1 is known to be expressed in the dental mesenchyme. However, little is known about how BMP signaling mediated by ACVR1 regulates dentinogenesis. To explore the role of ACVR1 in dentin formation in molars and incisors in mice, Acvr1 was conditionally disrupted in Osterix-expressing cells (designated as cKO). We found that loss of Acvr1 in the dental mesenchyme led to dentin dysplasia in molars and osteodentin formation in incisors. Specifically, the cKO mice exhibited remarkable tooth phenotypes characterized by thinner dentin and thicker predentin, as well as compromised differentiation of odontoblasts in molars. We also found osteodentin formation in the coronal part of the cKO mandibular incisors, which was associated with a reduction in the expression of odontogenic gene Dsp and an increase in the expression of osteogenic gene Bsp, leading to an alteration of cell fate from odontoblasts to osteoblasts. In addition, the expressions of WNT antagonists, Dkk1 and Sost, were downregulated and B-catenin was up-regulated in the cKO incisors, while the expression levels were not changed in the cKO molars, compared with the corresponding controls. Our results indicate the distinct and critical roles of ACVR1 between incisors and molars, which is associated with alterations in the WNT signaling related molecules. This study demonstrates for the first time the physiological roles of ACVR1 during dentinogenesis.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Ce Shi
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Huan Zhao
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Yijun Zhou
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Yue Hu
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Guangxing Yan
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Cangwei Liu
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Daowei Li
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Xinqing Hao
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109-1078, USA
| | - Qilin Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China. .,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.
| | - Hongchen Sun
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, 130021, China.
| |
Collapse
|
50
|
Gomez-Puerto MC, Iyengar PV, García de Vinuesa A, Ten Dijke P, Sanchez-Duffhues G. Bone morphogenetic protein receptor signal transduction in human disease. J Pathol 2018; 247:9-20. [PMID: 30246251 PMCID: PMC6587955 DOI: 10.1002/path.5170] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/03/2018] [Accepted: 09/13/2018] [Indexed: 12/23/2022]
Abstract
Bone morphogenetic proteins (BMPs) are secreted cytokines that were initially discovered on the basis of their ability to induce bone. Several decades of research have now established that these proteins function in a large variety of physiopathological processes. There are about 15 BMP family members, which signal via three transmembrane type II receptors and four transmembrane type I receptors. Mechanistically, BMP binding leads to phosphorylation of the type I receptor by the type II receptor. This activated heteromeric complex triggers intracellular signaling that is initiated by phosphorylation of receptor‐regulated SMAD1, 5, and 8 (also termed R‐SMADs). Activated R‐SMADs form heteromeric complexes with SMAD4, which engage in specific transcriptional responses. There is convergence along the signaling pathway and, besides the canonical SMAD pathway, BMP‐receptor activation can also induce non‐SMAD signaling. Each step in the pathway is fine‐tuned by positive and negative regulation and crosstalk with other signaling pathways. For example, ligand bioavailability for the receptor can be regulated by ligand‐binding proteins that sequester the ligand from interacting with receptors. Accessory co‐receptors, also known as BMP type III receptors, lack intrinsic enzymatic activity but enhance BMP signaling by presenting ligands to receptors. In this review, we discuss the role of BMP receptor signaling and how corruption of this pathway contributes to cardiovascular and musculoskeletal diseases and cancer. We describe pharmacological tools to interrogate the function of BMP receptor signaling in specific biological processes and focus on how these agents can be used as drugs to inhibit or activate the function of the receptor, thereby normalizing dysregulated BMP signaling. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Maria Catalina Gomez-Puerto
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Prasanna Vasudevan Iyengar
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Amaya García de Vinuesa
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|