1
|
Li J, Liu W, Wang T, Wang Y, Yang G, Chen J, Xu Y, Yang J. The mechanism of curcumin protecting against IL-1β-induced oxidative stress and inflammation in chondrocytes via the Bmp2/Smad5/Runx2 pathway. Cytotechnology 2025; 77:71. [PMID: 40028369 PMCID: PMC11871223 DOI: 10.1007/s10616-025-00731-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
A core role of chondrocyte survival/death has been suggested in the pathogenesis of osteoarthritis. We explored the underlying molecular mechanism of curcumin protecting against interleukin-1β (IL-1β)-induced chondrocyte injury via the bone morphogenetic protein 2 (Bmp2)/small mothers against decapentaplegic homolog 5 (Smad5)/runt-related transcription factor 2 (Runx2) pathway. Chondrocytes ATDC5 in vitro inflammatory model was established by IL-1β induction, and treated with curcumin, or Smad5 small interfering RNA. Levels of extracellular matrix (ECM) type II collagen (Col-II) and aggrecan, reactive oxygen species (ROS), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α) and IL-6 were determined by immunocytochemistry, kits and ELISA. Apoptosis and necrosis were assessed by Annexin V/PI and TUNEL. Matrix metalloproteinase 13 (MMP13), A disintegrin and metalloproteinase with thrombospondin 5 (ADAMTS5), Bmp2/Smad5/Runx2 expression and Smad5 phosphorylation levels were determined by qPCR and western blot. IL-1β-treated ATDC5 cells showed decreased Col-II, aggrecan in ECM and SOD and GSH-Px levels, as well as increased apoptosis and levels of MMP13, ADAMTS5, Bmp2, Runx2, ROS, COX-2, TNF-α and IL-6 and Smad5 phosphorylation (all p < 0.05), whilst curcumin treatment brought about the opposite trends, suggesting that curcumin inhibited oxidative stress, inflammatory response and apoptosis, and inactivated the Bmp2/Smad5/Runx2 pathway in IL-1β-treated chondrocytes. Additionally, Smad5 silencing also caused suppressed oxidative stress, inflammatory response and apoptosis in IL-1β-treated chondrocytes. Curcumin reduced IL-1β-induced chondrocyte oxidative stress, inflammation, and apoptosis, and increased ECM secretion by inactivating the Bmp2/Smad5/Runx2 pathway, thereby exerting a protective effect on injured chondrocytes.
Collapse
Affiliation(s)
- Jinlei Li
- Department of Orthopedics and Traumatology, Kunming Municipal Hospital of Traditional Chinese Medicine, No. 25, Dongfeng East Road, Panlong District, KunmingYunnan, 650600 China
| | - Weitong Liu
- Department of Orthopedics and Traumatology, Kunming Municipal Hospital of Traditional Chinese Medicine, No. 25, Dongfeng East Road, Panlong District, KunmingYunnan, 650600 China
| | - Tao Wang
- Department of Orthopedics and Traumatology, Kunming Municipal Hospital of Traditional Chinese Medicine, No. 25, Dongfeng East Road, Panlong District, KunmingYunnan, 650600 China
| | - Yanbo Wang
- Department of Orthopedics and Traumatology, Kunming Municipal Hospital of Traditional Chinese Medicine, No. 25, Dongfeng East Road, Panlong District, KunmingYunnan, 650600 China
| | - Guang Yang
- Department of Orthopedics and Traumatology, Kunming Municipal Hospital of Traditional Chinese Medicine, No. 25, Dongfeng East Road, Panlong District, KunmingYunnan, 650600 China
| | - Jiankun Chen
- Department of Orthopedics and Traumatology, Kunming Municipal Hospital of Traditional Chinese Medicine, No. 25, Dongfeng East Road, Panlong District, KunmingYunnan, 650600 China
| | - Yongsheng Xu
- Department of Orthopedics and Traumatology, Lincang People’s Hospital, Lincang, 677000 Yunnan China
| | - Jingfan Yang
- Department of Orthopedics and Traumatology, Kunming Municipal Hospital of Traditional Chinese Medicine, No. 25, Dongfeng East Road, Panlong District, KunmingYunnan, 650600 China
| |
Collapse
|
2
|
Lang A, Benn A, Collins JM, Wolter A, Balcaen T, Kerckhofs G, Zwijsen A, Boerckel JD. Endothelial SMAD1/5 signaling couples angiogenesis to osteogenesis in juvenile bone. Commun Biol 2024; 7:315. [PMID: 38480819 PMCID: PMC10937971 DOI: 10.1038/s42003-024-05915-1] [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: 02/22/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Skeletal development depends on coordinated angiogenesis and osteogenesis. Bone morphogenetic proteins direct bone formation in part by activating SMAD1/5 signaling in osteoblasts. However, the role of SMAD1/5 in skeletal endothelium is unknown. Here, we found that endothelial cell-conditional SMAD1/5 depletion in juvenile mice caused metaphyseal and diaphyseal hypervascularity, resulting in altered trabecular and cortical bone formation. SMAD1/5 depletion induced excessive sprouting and disrupting the morphology of the metaphyseal vessels, with impaired anastomotic loop formation at the chondro-osseous junction. Endothelial SMAD1/5 depletion impaired growth plate resorption and, upon long-term depletion, abrogated osteoprogenitor recruitment to the primary spongiosa. Finally, in the diaphysis, endothelial SMAD1/5 activity was necessary to maintain the sinusoidal phenotype, with SMAD1/5 depletion inducing formation of large vascular loops and elevated vascular permeability. Together, endothelial SMAD1/5 activity sustains skeletal vascular morphogenesis and function and coordinates growth plate remodeling and osteoprogenitor recruitment dynamics in juvenile mouse bone.
Collapse
Affiliation(s)
- Annemarie Lang
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany.
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden (TUD), Fetscherstrasse 74, Dresden, 01307, Germany.
| | - Andreas Benn
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, 3000, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Leuven, 3000, Belgium
| | - Joseph M Collins
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Angelique Wolter
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, 14163, Germany
| | - Tim Balcaen
- Institute of Mechanics, Materials and Civil Engineering, Biomechanics lab, UCLouvain, Louvain-la-Neuve, 1348, Belgium
- Institute of Experimental and Clinical Research, Pole of Morphology, UCLouvain, Brussels, 1200, Belgium
- KU Leuven, Department of Chemistry, Sustainable Chemistry for Metals and Molecules, Leuven, 3000, Belgium
| | - Greet Kerckhofs
- Institute of Mechanics, Materials and Civil Engineering, Biomechanics lab, UCLouvain, Louvain-la-Neuve, 1348, Belgium
- Institute of Experimental and Clinical Research, Pole of Morphology, UCLouvain, Brussels, 1200, Belgium
- Department of Materials Engineering, KU Leuven, Heverlee, 3001, Belgium
- Division for Skeletal Tissue Engineering, Prometheus, KU Leuven, Leuven, 3000, Belgium
| | - An Zwijsen
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, 3000, Belgium
| | - Joel D Boerckel
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
3
|
Chen S, Wang F, Yang G, Yuan G, Liu M, Goldman G, Harris S, Wang W, Chen Z, Mary M. Loss of Bmp2 impairs odontogenesis via dysregulating pAkt/pErk/GCN5/Dlx3/Sp7. RESEARCH SQUARE 2023:rs.3.rs-3299295. [PMID: 37790473 PMCID: PMC10543288 DOI: 10.21203/rs.3.rs-3299295/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
BMP2 signaling plays a pivotal role in odontoblast differentiation and maturation during odontogenesis. Teeth lacking Bmp2 exhibit a morphology reminiscent of dentinogenesis imperfecta (DGI), associated with mutations in dentin matrix protein 1 (DMP1) and dentin sialophosphoprotein (DSPP) genes. Mechanisms by which BMP2 signaling influences expressions of DSPP and DMP1 and contributes to DGI remain elusive. To study the roles of BMP2 in dentin development, we generated Bmp2 conditional knockout (cKO) mice. Through a comprehensive approach involving RNA-seq, immunohistochemistry, promoter activity, ChIP, and Re-ChIP, we investigated downstream targets of Bmp2. Notably, the absence of Bmp2 in cKO mice led to dentin insufficiency akin to DGI. Disrupted Bmp2 signaling was linked to decreased expression of Dspp and Dmp1, as well as alterations in intracellular translocation of transcription factors Dlx3 and Sp7. Intriguingly, upregulation of Dlx3, Dmp1, Dspp, and Sp7, driven by BMP2, fostered differentiation of dental mesenchymal cells and biomineralization. Mechanistically, BMP2 induced phosphorylation of Dlx3, Sp7, and histone acetyltransferase GCN5 at Thr and Tyr residues, mediated by Akt and Erk42/44 kinases. This phosphorylation facilitated protein nuclear translocation, promoting interactions between Sp7 and Dlx3, as well as with GCN5 on Dspp and Dmp1 promoters. The synergy between Dlx3 and Sp7 bolstered transcription of Dspp and Dmp1. Notably, BMP2-driven GCN5 acetylated Sp7 and histone H3, while also recruiting RNA polymerase II to Dmp1 and Dspp chromatins, enhancing their transcriptions. Intriguingly, BMP2 suppressed the expression of histone deacetylases. we unveil hitherto uncharted involvement of BMP2 in dental cell differentiation and dentine development through pAkt/pErk42/44/Dlx3/Sp7/GCN5/Dspp/Dmp1.
Collapse
Affiliation(s)
- Shuo Chen
- UT Health Science Center at San Antonio
| | | | | | | | - Mengmeng Liu
- School of Dentistry, the University of Texas Health Science Center at San Antonio
| | - Graham Goldman
- School of Dentistry, the University of Texas Health Science Center at San Antonio
| | | | | | - Zhi Chen
- Wuhan University School and Hospital of Stomatology
| | | |
Collapse
|
4
|
Rikitake K, Kunimatsu R, Yoshimi Y, Nakajima K, Hiraki T, Aisyah Rizky Putranti N, Tsuka Y, Abe T, Ando K, Hayashi Y, Nikawa H, Tanimoto K. Effect of CD146 + SHED on bone regeneration in a mouse calvaria defect model. Oral Dis 2023; 29:725-734. [PMID: 34510661 DOI: 10.1111/odi.14020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Stem cells from human exfoliated deciduous teeth (SHED) have bone regeneration ability and potential therapeutic applications. CD146, a cell adhesion protein expressed by vascular endothelial cells, is involved in osteoblastic differentiation of stem cells. The effect of CD146 on SHED-mediated bone regeneration in vivo remains unknown. We aimed to establish efficient conditions for SHED transplantation. MATERIALS AND METHODS SHED were isolated from the pulp of an extracted deciduous tooth and cultured; CD146-positive (CD146+ ) and CD146-negative (CD146- ) populations were sorted. Heterogeneous populations of SHED and CD146+ and CD146- cells were transplanted into bone defects generated in the skulls of immunodeficient mice. Micro-computed tomography was performed immediately and 4 and 8 weeks later. Histological and immunohistochemical assessments were performed 8 weeks later. RESULTS Bone regeneration was observed upon transplantation with CD146+ and heterogeneous populations of SHED, with significantly higher bone regeneration observed with CD146+ cells. Bone regeneration was higher in the CD146- group than in the control group, but significantly lower than that in the other transplant groups at 4 and 8 weeks. Histological and immunohistochemical assessments revealed that CD146+ cells promoted bone regeneration and angiogenesis. CONCLUSION Transplantation of CD146+ SHED into bone defects may be useful for bone regeneration.
Collapse
Affiliation(s)
- Kodai Rikitake
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Ryo Kunimatsu
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuki Yoshimi
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kengo Nakajima
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Tomoka Hiraki
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Nurul Aisyah Rizky Putranti
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuji Tsuka
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takaharu Abe
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kazuyo Ando
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yoko Hayashi
- Analysis Center of Life Science, Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Hiroki Nikawa
- Department of Oral Biology and Engineering, Division of Oral Health Sciences, Institute of Biomedical and Health Sciences, Hiroshima University Graduate School, Hiroshima, Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
5
|
Lang A, Benn A, Wolter A, Balcaen T, Collins J, Kerckhofs G, Zwijsen A, Boerckel JD. Endothelial SMAD1/5 signaling couples angiogenesis to osteogenesis during long bone growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.522994. [PMID: 36712097 PMCID: PMC9881901 DOI: 10.1101/2023.01.07.522994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Skeletal development depends on coordinated angiogenesis and osteogenesis. Bone morphogenetic proteins direct bone development by activating SMAD1/5 signaling in osteoblasts. However, the role of SMAD1/5 in skeletal endothelium is unknown. Here, we found that endothelial cell-conditional SMAD1/5 depletion in juvenile mice caused metaphyseal and diaphyseal hypervascularity, resulting in altered cancellous and cortical bone formation. SMAD1/5 depletion induced excessive sprouting, disrupting the columnar structure of the metaphyseal vessels and impaired anastomotic loop morphogenesis at the chondro-osseous junction. Endothelial SMAD1/5 depletion impaired growth plate resorption and, upon long term depletion, abrogated osteoprogenitor recruitment to the primary spongiosa. Finally, in the diaphysis, endothelial SMAD1/5 activity was necessary to maintain the sinusoidal phenotype, with SMAD1/5 depletion inducing formation of large vascular loops, featuring elevated endomucin expression, ectopic tip cell formation, and hyperpermeability. Together, endothelial SMAD1/5 activity sustains skeletal vascular morphogenesis and function and coordinates growth plate remodeling and osteoprogenitor recruitment dynamics during bone growth.
Collapse
Affiliation(s)
- Annemarie Lang
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - Andreas Benn
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Angelique Wolter
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Tim Balcaen
- Biomechanics lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Molecular Design and Synthesis, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Joseph Collins
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Materials Engineering, KU Leuven, Heverlee, Belgium
- Prometheus, Division for Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - An Zwijsen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Belgium
| | - Joel D. Boerckel
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
6
|
Juuri E, Tikka P, Domanskyi A, Corfe I, Morita W, Mckinnon PJ, Jandova N, Balic A. Ptch2 is a Potential Regulator of Mesenchymal Stem Cells. Front Physiol 2022; 13:877565. [PMID: 35574464 PMCID: PMC9096555 DOI: 10.3389/fphys.2022.877565] [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: 02/16/2022] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Ptch receptors 1 and 2 mediate Hedgehog signaling pivotal for organ development and homeostasis. In contrast to embryonic lethal Ptch1−/− phenotype, Ptch2−/− mice display no effect on gross phenotype. In this brief report, we provide evidence of changes in the putative incisor mesenchymal stem cell (MSC) niches that contribute to accelerated incisor growth, as well as intriguing changes in the bones and skin which suggest a role for Ptch2 in the regulation of MSCs and their regenerative potential. We employed histological, immunostaining, and computed tomography (µCT) analyses to analyze morphological differences between Ptch2−/− and wild-type incisors, long bones, and skins. In vitro CFU and differentiation assays were used to demonstrate the MSC content and differentiation potential of Ptch2−/− bone marrow stromal cells. Wound healing assay was performed in vivo and in vitro on 8-week-old mice to assess the effect of Ptch2 on the wound closure. Loss of Ptch2 causes increases in the number of putative MSCs in the continuously growing incisor, associated with increased vascularization observed in the tooth mesenchyme and the neurovascular bundle. Increased length and volume of Ptch2−/− bones is linked with the increased number and augmented in vitro differentiation potential of MSCs in the bone marrow. Dynamic changes in the Ptch2−/− skin thickness relate to changes in the mesenchymal compartment and impact the wound closure potential. The effects of Ptch2 abrogation on the postnatal MSCs suggest a crucial role for Ptch2 in Hedgehog signaling regulation of the organ regenerative potential.
Collapse
Affiliation(s)
- Emma Juuri
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Oral and Maxillofacial Diseases, Helsinki University Hospital, Helsinki, Finland
| | - Pauli Tikka
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Andrii Domanskyi
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ian Corfe
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Circuar Economy Solutions Unit, Geological Survey of Finland, Espoo, Finland
| | - Wataru Morita
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Department of Anthropology, National Museum of Nature and Science, Taito, Japan
| | - Peter J Mckinnon
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Nela Jandova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia.,Institute of Animal Physiology and Genetics, CAS, Brno, Czechia
| | - Anamaria Balic
- Cell and Tissue Dynamics Research Program, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Institute of Oral Biology, Centre for Dental Medicine, University of Zürich, Zürich, Switzerland
| |
Collapse
|
7
|
Nanohydroxyapatite-Blasted Bioactive Surface Drives Shear-Stressed Endothelial Cell Growth and Angiogenesis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1433221. [PMID: 35252440 PMCID: PMC8890866 DOI: 10.1155/2022/1433221] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 12/22/2022]
Abstract
Nanosized crystalline hydroxyapatite coating (HAnano®) accelerates the osteointegration of dental implants which is hypothesized to drive angiogenesis. In order to test this hypothesis, we have subjected shear-stressed human umbilical vein endothelial cells (HUVECs) to a HAnano®-enriched medium, as well as to surface presenting dual acid etching (DAE) as a control. To note, the titanium implants were coated with 10 nm in diameter HA particles using the Promimic HAnano method. Our data reveals that HAnano® modulates higher expression of genes related with endothelial cell performance and viability, such as VEGF, eNOS, and AKT, and further angiogenesis in vitro by promoting endothelial cell migration. Additionally, the data shows a significant extracellular matrix (ECM) remodeling, and this finding seems developing a dual role in promoting the expression of VEGF and control endothelial cell growth during angiogenesis. Altogether, these data prompted us to further validate this phenomenon by exploring genes related with the control of cell cycle and in fact our data shows that HAnano® promotes higher expression of CDK4 gene, while p21 and p15 genes (suppressor genes) were significantly lower. In conjunction, our data shows for the first time that HAnano®-coated surfaces drive angiogenesis by stimulating a proliferative and migration phenotype of endothelial cells, and this finding opens novel comprehension about osseointegration mechanism considering nanosized hydroxyapatite coating dental implants.
Collapse
|
8
|
Mechanical regulation of bone remodeling. Bone Res 2022; 10:16. [PMID: 35181672 PMCID: PMC8857305 DOI: 10.1038/s41413-022-00190-4] [Citation(s) in RCA: 215] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022] Open
Abstract
Bone remodeling is a lifelong process that gives rise to a mature, dynamic bone structure via a balance between bone formation by osteoblasts and resorption by osteoclasts. These opposite processes allow the accommodation of bones to dynamic mechanical forces, altering bone mass in response to changing conditions. Mechanical forces are indispensable for bone homeostasis; skeletal formation, resorption, and adaptation are dependent on mechanical signals, and loss of mechanical stimulation can therefore significantly weaken the bone structure, causing disuse osteoporosis and increasing the risk of fracture. The exact mechanisms by which the body senses and transduces mechanical forces to regulate bone remodeling have long been an active area of study among researchers and clinicians. Such research will lead to a deeper understanding of bone disorders and identify new strategies for skeletal rejuvenation. Here, we will discuss the mechanical properties, mechanosensitive cell populations, and mechanotransducive signaling pathways of the skeletal system.
Collapse
|
9
|
Toth Z, Ward A, Tang SY, McBride-Gagyi S. Sexual differences in bone porosity, osteocyte density, and extracellular matrix organization due to osteoblastic-specific Bmp2 deficiency in mice. Bone 2021; 150:116002. [PMID: 33971313 PMCID: PMC8217247 DOI: 10.1016/j.bone.2021.116002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/01/2022]
Abstract
Clinical studies have come to conflicting conclusions regarding BMP2 deficiency's link to regulating bone mass and increasing fracture risk. This may be due to the signaling protein having sex- or age-dependent effects. Previous pre-clinical studies have supported a role, but have not adequately determined the physical mechanism causing altered bulk material properties. This study investigated the physical effects of Bmp2 ablation from osteogenic lineage cells (Osx-Cre; Bmp2fl/fl) in 10- and 15-week-old male and female mice. Bones collected post-mortem were subjected to fracture toughness testing, reference point indentation testing, microCT, and histological analysis to determine the multi-scale relationships between mechanical/material behavior and collagen production, collagen organization, and bone architecture. BMP2-deficient bones were smaller, more brittle, and contained more lacunae-scale voids and cortical pores. The cellular density was significantly increased and there were material-level differences measured by reference point indentation, independently of collagen fiber alignment or organization. The disparities in bone size and in bone fracture toughness between genotypes were especially striking in males at 15-weeks-old. Together, this study suggests that there are sex- and age-dependent effects of BMP2 deficiency. The results from both sexes also warrant further investigation into BMP2 deficiency's role in osteoblasts' transition to osteocytes and overall bone porosity.
Collapse
Affiliation(s)
- Zacharie Toth
- Department of Orthopaedic Surgery, Saint Louis University, St. Louis, MO, United States of America
| | - Ashley Ward
- Department of Orthopaedic Surgery, Saint Louis University, St. Louis, MO, United States of America
| | - Simon Y Tang
- Department of Orthopaedics, Washington University in St. Louis, St. Louis, MO, United States of America
| | - Sarah McBride-Gagyi
- Department of Orthopaedic Surgery, Saint Louis University, St. Louis, MO, United States of America.
| |
Collapse
|
10
|
Zhang N, Cui M, Liu X, Yu L, Zhao X, Cao L, Ji Y. IL-17F promotes osteoblastic osteogenesis via the MAPK/ERK1/2 signaling pathway. Exp Ther Med 2021; 22:1052. [PMID: 34434266 PMCID: PMC8353634 DOI: 10.3892/etm.2021.10486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/23/2020] [Indexed: 12/27/2022] Open
Abstract
Osteoimmunology is a field that focuses on the interactions between the skeletal and immune systems, and has become a focus of research over the years. The role of interleukin (IL)-17F, a proinflammatory cytokine, in bone regeneration and its signal transduction are not completely understood. The aim of the present study was to evaluate the function of IL-17F and the possible mechanisms underlying IL-17F in osteoblasts in vitro. Osteoblasts derived from newborn rats were treated with various concentrations of IL-17F. The pro-osteogenic effects of IL-17F were assessed at the cellular and molecular level. The results demonstrated that IL-17F promoted osteoblast proliferation, differentiation and mineralization. Reverse transcription-quantitative PCR and western blotting indicated that IL-17F treatment upregulated osteogenesis-related factors, including bone morphogenetic protein-2, Runt-related transcription factor-2 (Runx2) and Osterix, and downregulated Noggin compared with the control group. Subsequently, whether the IL-17F receptors, IL-17 receptor (IL-17R) A and IL-17RC, served a role in the effects of IL-17F on osteoblasts was investigated. The mRNA expression levels of IL-17RA and IL-17RC were upregulated in IL-17F-treated osteoblasts compared with control osteoblasts. Furthermore, U0126, a MAPK/ERK1/2 inhibitor, was utilized to investigate the mechanisms underlying IL-17F. The results indicated that compared with the control group, IL-17F increased the protein expression of phosphorylated-ERK1/2, Runx2 and Osterix, whereas U0126 reversed IL-17F-mediated effects. Collectively, the results of the present study suggested that IL-17F promoted osteoblastic osteogenesis via the MAPK/ERK1/2-mediated signaling pathway. IL-17F promoted osteogenesis, including proliferation, differentiation and mineralization activity, indicating that IL-17F may serve as a potential therapeutic target for osteoblast-mediated bone loss disease.
Collapse
Affiliation(s)
- Na Zhang
- Department of Pain Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Min Cui
- Department of Pain Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Xudong Liu
- Department of Pain Medicine, Shandong University Qilu Hospital, Jinan, Shandong 250012, P.R. China
| | - Lingzhi Yu
- Department of Pain Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, P.R. China
| | - Xu Zhao
- Department of Anesthesiology, Shandong Provincial Hospital, Jinan, Shandong 250021, P.R. China
| | - Luning Cao
- Department of Pain Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272001, P.R. China
| | - Yuanyuan Ji
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
11
|
Chen M, Li Y, Huang X, Gu Y, Li S, Yin P, Zhang L, Tang P. Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res 2021; 9:21. [PMID: 33753717 PMCID: PMC7985324 DOI: 10.1038/s41413-021-00138-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis and osteogenesis are coupled. However, the cellular and molecular regulation of these processes remains to be further investigated. Both tissues have recently been recognized as endocrine organs, which has stimulated research interest in the screening and functional identification of novel paracrine factors from both tissues. This review aims to elaborate on the novelty and significance of endocrine regulatory loops between bone and the vasculature. In addition, research progress related to the bone vasculature, vessel-related skeletal diseases, pathological conditions, and angiogenesis-targeted therapeutic strategies are also summarized. With respect to future perspectives, new techniques such as single-cell sequencing, which can be used to show the cellular diversity and plasticity of both tissues, are facilitating progress in this field. Moreover, extracellular vesicle-mediated nuclear acid communication deserves further investigation. In conclusion, a deeper understanding of the cellular and molecular regulation of angiogenesis and osteogenesis coupling may offer an opportunity to identify new therapeutic targets.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ya Gu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Shang Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| |
Collapse
|
12
|
Yu S, Guo J, Sun Z, Lin C, Tao H, Zhang Q, Cui Y, Zuo H, Lin Y, Chen S, Liu H, Chen Z. BMP2-dependent gene regulatory network analysis reveals Klf4 as a novel transcription factor of osteoblast differentiation. Cell Death Dis 2021; 12:197. [PMID: 33608506 PMCID: PMC7895980 DOI: 10.1038/s41419-021-03480-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
Transcription factors (TFs) regulate the expression of target genes, inducing changes in cell morphology or activities needed for cell fate determination and differentiation. The BMP signaling pathway is widely regarded as one of the most important pathways in vertebrate skeletal biology, of which BMP2 is a potent inducer, governing the osteoblast differentiation of bone marrow stromal cells (BMSCs). However, the mechanism by which BMP2 initiates its downstream transcription factor cascade and determines the direction of differentiation remains largely unknown. In this study, we used RNA-seq, ATAC-seq, and animal models to characterize the BMP2-dependent gene regulatory network governing osteoblast lineage commitment. Sp7-Cre; Bmp2fx/fx mice (BMP2-cKO) were generated and exhibited decreased bone density and lower osteoblast number (n > 6). In vitro experiments showed that BMP2-cKO mouse bone marrow stromal cells (mBMSCs) had an impact on osteoblast differentiation and deficient cell proliferation. Osteogenic medium induced mBMSCs from BMP2-cKO mice and control were subjected to RNA-seq and ATAC-seq analysis to reveal differentially expressed TFs, along with their target open chromatin regions. Combined with H3K27Ac CUT&Tag during osteoblast differentiation, we identified 2338 BMP2-dependent osteoblast-specific active enhancers. Motif enrichment assay revealed that over 80% of these elements were directly targeted by RUNX2, DLX5, MEF2C, OASIS, and KLF4. We deactivated Klf4 in the Sp7 + lineage to validate the role of KLF4 in osteoblast differentiation of mBMSCs. Compared to the wild-type, Sp7-Cre; Klf4fx/+ mice (KLF4-Het) were smaller in size and had abnormal incisors resembling BMP2-cKO mice. Additionally, KLF4-Het mice had fewer osteoblasts and decreased osteogenic ability. RNA-seq and ATAC-seq revealed that KLF4 mainly "co-bound" with RUNX2 to regulate downstream genes. Given the significant overlap between KLF4- and BMP2-dependent NFRs and enriched motifs, our findings outline a comprehensive BMP2-dependent gene regulatory network specifically governing osteoblast differentiation of the Sp7 + lineage, in which Klf4 is a novel transcription factor.
Collapse
Affiliation(s)
- Shuaitong Yu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jinqiang Guo
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zheyi Sun
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chujiao Lin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huangheng Tao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qian Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu Cui
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huanyan Zuo
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yuxiu Lin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shuo Chen
- Department of Developmental Dentistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Huan Liu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhi Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Chiou G, Jui E, Rhea AC, Gorthi A, Miar S, Acosta FM, Perez C, Suhail Y, Kshitiz, Chen Y, Ong JL, Bizios R, Rathbone C, Guda T. Scaffold Architecture and Matrix Strain Modulate Mesenchymal Cell and Microvascular Growth and Development in a Time Dependent Manner. Cell Mol Bioeng 2020; 13:507-526. [PMID: 33184580 PMCID: PMC7596170 DOI: 10.1007/s12195-020-00648-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 08/11/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Volumetric tissue-engineered constructs are limited in development due to the dependence on well-formed vascular networks. Scaffold pore size and the mechanical properties of the matrix dictates cell attachment, proliferation and successive tissue morphogenesis. We hypothesize scaffold pore architecture also controls stromal-vessel interactions during morphogenesis. METHODS The interaction between mesenchymal stem cells (MSCs) seeded on hydroxyapatite scaffolds of 450, 340, and 250 μm pores and microvascular fragments (MVFs) seeded within 20 mg/mL fibrin hydrogels that were cast into the cell-seeded scaffolds, was assessed in vitro over 21 days and compared to the fibrin hydrogels without scaffold but containing both MSCs and MVFs. mRNA sequencing was performed across all groups and a computational mechanics model was developed to validate architecture effects on predicting vascularization driven by stiffer matrix behavior at scaffold surfaces compared to the pore interior. RESULTS Lectin staining of decalcified scaffolds showed continued vessel growth, branching and network formation at 14 days. The fibrin gel provides no resistance to spread-out capillary networks formation, with greater vessel loops within the 450 μm pores and vessels bridging across 250 μm pores. Vessel growth in the scaffolds was observed to be stimulated by hypoxia and successive angiogenic signaling. Fibrin gels showed linear fold increase in VEGF expression and no change in BMP2. Within scaffolds, there was multiple fold increase in VEGF between days 7 and 14 and early multiple fold increases in BMP2 between days 3 and 7, relative to fibrin. There was evidence of yap/taz based hippo signaling and mechanotransduction in the scaffold groups. The vessel growth models determined by computational modeling matched the trends observed experimentally. CONCLUSION The differing nature of hypoxia signaling between scaffold systems and mechano-transduction sensing matrix mechanics were primarily responsible for differences in osteogenic cell and microvessel growth. The computational model implicated scaffold architecture in dictating branching morphology and strain in the hydrogel within pores in dictating vessel lengths.
Collapse
Affiliation(s)
- Gennifer Chiou
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Elysa Jui
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Allison C. Rhea
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Aparna Gorthi
- Greehey Children’s Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX 78229 USA
| | - Solaleh Miar
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Francisca M. Acosta
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Cynthia Perez
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030 USA
- Cancer Systems Biology at Yale, Yale University, West Haven, CT 06516 USA
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX 78229 USA
| | - Joo L. Ong
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Rena Bizios
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Christopher Rathbone
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249 USA
| |
Collapse
|
14
|
Mang T, Kleinschmidt-Doerr K, Ploeger F, Schoenemann A, Lindemann S, Gigout A. BMPR1A is necessary for chondrogenesis and osteogenesis, whereas BMPR1B prevents hypertrophic differentiation. J Cell Sci 2020; 133:jcs246934. [PMID: 32764110 DOI: 10.1242/jcs.246934] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/21/2020] [Indexed: 08/31/2023] Open
Abstract
BMP2 stimulates bone formation and signals preferably through BMP receptor (BMPR) 1A, whereas GDF5 is a cartilage inducer and signals preferably through BMPR1B. Consequently, BMPR1A and BMPR1B are believed to be involved in bone and cartilage formation, respectively. However, their function is not yet fully clarified. In this study, GDF5 mutants with a decreased affinity for BMPR1A were generated. These mutants, and wild-type GDF5 and BMP2, were tested for their ability to induce dimerization of BMPR1A or BMPR1B with BMPR2, and for their chondrogenic, hypertrophic and osteogenic properties in chondrocytes, in the multipotent mesenchymal precursor cell line C3H10T1/2 and the human osteosarcoma cell line Saos-2. Mutants with the lowest potency for inducing BMPR1A-BMPR2 dimerization exhibited minimal chondrogenic and osteogenic activities, indicating that BMPR1A is necessary for chondrogenic and osteogenic differentiation. BMP2, GDF5 and the GDF5 R399E mutant stimulated expression of chondrogenic and hypertrophy markers in C3H10T1/2 cells and chondrocytes. However, GDF5 R399E, which induces the dimerization of BMPR1B and BMPR2 more potently than GDF5 or BMP2, displayed reduced hypertrophic activity. Therefore, we postulate that stronger BMPR1B signaling, compared to BMPR1A signaling, prevents chondrocyte hypertrophy and acts as a cartilage stabilizer during joint morphogenesis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tanja Mang
- Osteoarthritis Research, Merck KGaA, 64293 Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität, 64289 Darmstadt, Germany
| | | | | | | | - Sven Lindemann
- Osteoarthritis Research, Merck KGaA, 64293 Darmstadt, Germany
| | - Anne Gigout
- Osteoarthritis Research, Merck KGaA, 64293 Darmstadt, Germany
| |
Collapse
|
15
|
Sears C, Mondragon E, Richards ZI, Sears N, Chimene D, McNeill EP, Gregory CA, Gaharwar AK, Kaunas R. Conditioning of 3D Printed Nanoengineered Ionic-Covalent Entanglement Scaffolds with iP-hMSCs Derived Matrix. Adv Healthc Mater 2020; 9:e1901580. [PMID: 32147960 PMCID: PMC7500865 DOI: 10.1002/adhm.201901580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/30/2020] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
Additive manufacturing is a promising method for producing customized 3D bioactive constructs for regenerative medicine. Here, 3D printed highly osteogenic scaffolds using nanoengineered ionic-covalent entanglement ink (NICE) for bone tissue engineering are reported. This NICE ink consists of ionic-covalent entanglement reinforced with Laponite, a 2D nanosilicate (nSi) clay, allowing for the printing of anatomic-sized constructs with high accuracy. The 3D printed structure is able to maintain high structural stability in physiological conditions without any significant swelling or deswelling. The presence of nSi imparts osteoinductive characteristics to the NICE scaffolds, which is further augmented by depositing pluripotent stem cell-derived extracellular matrix (ECM) on the scaffolds. This is achieved by stimulating human induced pluripotent stem cell-derived mesenchymal stem cells (iP-hMSCs) with 2-chloro-5-nitrobenzanilide, a PPARγ inhibitor that enhances Wnt pathway, resulting in the deposition of an ECM characterized by high levels of collagens VI and XII found in anabolic bone. The osteoinductive characteristics of these bioconditioned NICE (bNICE) scaffolds is demonstrated through osteogenic differentiation of bone marrow derived human mesenchymal stem cells. A significant increase in the expression of osteogenic gene markers as well as mineralized ECM are observed on bioconditioned NICE (bNICE) scaffolds compared to bare scaffolds (NICE). The bioconditioned 3D printed scaffolds provide a unique strategy to design personalized bone grafts for in situ bone regeneration.
Collapse
Affiliation(s)
- Candice Sears
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Eli Mondragon
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Zachary I Richards
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Nick Sears
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - David Chimene
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Eoin P McNeill
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Bryan, TX, 77807, USA
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Bryan, TX, 77807, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, TX, 77843, USA
| | - Roland Kaunas
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, 77843, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Bryan, TX, 77807, USA
| |
Collapse
|
16
|
Hashimoto K, Kaito T, Furuya M, Seno S, Okuzaki D, Kikuta J, Tsukazaki H, Matsuda H, Yoshikawa H, Ishii M. In vivo dynamic analysis of BMP-2-induced ectopic bone formation. Sci Rep 2020; 10:4751. [PMID: 32179857 PMCID: PMC7076033 DOI: 10.1038/s41598-020-61825-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/03/2020] [Indexed: 12/29/2022] Open
Abstract
Bone morphogenetic protein (BMP)-2 plays a central role in bone-tissue engineering because of its potent bone-induction ability. However, the process of BMP-induced bone formation in vivo remains poorly elucidated. Here, we aimed to establish a method for intravital imaging of the entire process of BMP-2-induced ectopic bone formation. Using multicolor intravital imaging in transgenic mice, we visualized the spatiotemporal process of bone induction, including appearance and motility of osteoblasts and osteoclasts, angiogenesis, collagen-fiber formation, and bone-mineral deposition. Furthermore, we investigated how PTH1-34 affects BMP-2-induced bone formation, which revealed that PTH1-34 administration accelerated differentiation and increased the motility of osteoblasts, whereas it decreased morphological changes in osteoclasts. This is the first report on visualization of the entire process of BMP-2-induced bone formation using intravital imaging techniques, which, we believe, will contribute to our understanding of ectopic bone formation and provide new parameters for evaluating bone-forming activity.
Collapse
Affiliation(s)
- Kunihiko Hashimoto
- Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan.
| | - Masayuki Furuya
- Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan.,Department of Orthopaedic Surgery, Japan Organization of Occupational Health and Safety Osaka Rosai Hospital, Osaka, 591-8025, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science & Technology, Osaka University, Osaka, 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan
| | - Hiroyuki Tsukazaki
- Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science & Technology, Osaka University, Osaka, 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
17
|
Sharma A, Goring A, Staines KA, Emery RJ, Pitsillides AA, Oreffo RO, Mahajan S, Clarkin CE. Raman spectroscopy links differentiating osteoblast matrix signatures to pro-angiogenic potential. Matrix Biol Plus 2020; 5:100018. [PMID: 33543015 PMCID: PMC7852201 DOI: 10.1016/j.mbplus.2019.100018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/21/2019] [Accepted: 10/09/2019] [Indexed: 01/25/2023] Open
Abstract
Mineralization of bone is achieved by the sequential maturation of the immature amorphous calcium phase to mature hydroxyapatite (HA) and is central in the process of bone development and repair. To study normal and dysregulated mineralization in vitro, substrates are often coated with poly-l-lysine (PLL) which facilitates cell attachment. This study has used Raman spectroscopy to investigate the effect of PLL coating on osteoblast (OB) matrix composition during differentiation, with a focus on collagen specific proline and hydroxyproline and precursors of HA. Deconvolution analysis of murine derived long bone OB Raman spectra revealed collagen species were 4.01-fold higher in OBs grown on PLL. Further, an increase of 1.91-fold in immature mineral species (amorphous calcium phosphate) was coupled with a 9.32-fold reduction in mature mineral species (carbonated apatite) on PLL versus controls. These unique low mineral signatures identified in OBs were linked with reduced alkaline phosphatase enzymatic activity, reduced Alizarin Red staining and altered osteogenic gene expression. The promotion of immature mineral species and restriction of mature mineral species of OB grown on PLL were linked to increased cell viability and pro-angiogenic vascular endothelial growth factor (VEGF) production. These results demonstrate the utility of Raman spectroscopy to link distinct matrix signatures with OB maturation and VEGF release. Importantly, Raman spectroscopy could provide a label-free approach to clinically assess the angiogenic potential of bone during fracture repair or degenerative bone loss.
Collapse
Key Words
- ACP, amorphous calcium phosphate
- ALP, tissue non-specific alkaline phosphatase
- CAP, carbonated apatite
- CCEC, collagenase-collagenase-EDTA-collagenase
- ECM, extracellular matrix
- HA, hydroxyapatite
- HBSS, Hank's balanced salt solution
- MV, matrix vesicles
- OB, osteoblast
- OCP, octacalcium phosphate
- Osteoblast mineralization
- PCA, principle component analysis
- PLL, poly-l-lysine
- Poly-l-lysine
- RT-qPCR, reverse transcription-quantiative PCR
- Raman spectroscopy
- VEGF
- VEGF, vascular endothelial growth factor
Collapse
Affiliation(s)
- Aikta Sharma
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| | - Alice Goring
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| | - Katherine A. Staines
- School of Applied Sciences, Sighthill Campus, Edinburgh Napier University, Edinburgh, EH11 4BN, United Kingdom of Great Britain and Northern Ireland
| | - Roger J.H. Emery
- Department of Surgery and Cancer, Faculty of Medicine, St Mary's Campus, Imperial College London, London, W2 1PG, United Kingdom of Great Britain and Northern Ireland
| | - Andrew A. Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW1 0TU, United Kingdom of Great Britain and Northern Ireland
| | - Richard O.C. Oreffo
- Centre for Human Development, Stem Cell and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, United Kingdom of Great Britain and Northern Ireland
| | - Sumeet Mahajan
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| | - Claire E. Clarkin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton, SO17 1BJ, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
18
|
Urbano N, Scimeca M, Tancredi V, Bonanno E, Schillaci O. 99mTC-sestamibi breast imaging: Current status, new ideas and future perspectives. Semin Cancer Biol 2020; 84:302-309. [PMID: 31982511 DOI: 10.1016/j.semcancer.2020.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Here we proposed the most recent innovations in the use of Breast Specific Gamma Imaging with 99mTc-sestamibi for the management of breast cancer patients. To this end, we reported the recent discoveries concerning: a) the implementation of both instrumental devices and software, b) the biological mechanisms involved in the 99mTc-sestamibi uptake in breast cancer cells, c) the evaluation of Breast Specific Gamma Imaging with 99mTc-sestamibi as predictive markers of metastatic diseases. In this last case, we also reported preliminary data about the capability of Breast Specific Gamma Imaging with 99mTc-sestamibi to identify breast cancer lesions with high propensity to form bone metastatic lesions due to the presence of Breast Osteoblast-Like Cells.
Collapse
Affiliation(s)
- Nicoletta Urbano
- Nuclear Medicine, Policlinico "Tor Vergata", Viale Oxford, 81, 00133, Rome, Italy
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy; University of San Raffaele, Via di Val Cannuta 247, 00166, Rome, Italy; Fondazione Umberto Veronesi (FUV), Piazza Velasca 5, 20122, Milano (Mi), Italy; UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, School of Sport and Exercise Sciences, University of Rome Tor Vergata, Rome, Italy; Centre of Space Biomedicine, University of Rome Tor Vergata, Rome, Italy
| | - Elena Bonanno
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier, 1, 00133, Rome, Italy; Diagnostica Medica' & 'Villa dei Platani', Neuromed Group, Avellino, 83100, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy; IRCCS Neuromed, Pozzilli (Is), 86077, Italy.
| |
Collapse
|
19
|
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]
|
20
|
Qiu WX, Ma XL, Lin X, Zhao F, Li DJ, Chen ZH, Zhang KW, Zhang R, Wang P, Xiao YY, Miao ZP, Dang K, Wu XY, Qian AR. Deficiency of Macf1 in osterix expressing cells decreases bone formation by Bmp2/Smad/Runx2 pathway. J Cell Mol Med 2019; 24:317-327. [PMID: 31709715 PMCID: PMC6933318 DOI: 10.1111/jcmm.14729] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/16/2019] [Accepted: 10/03/2019] [Indexed: 12/18/2022] Open
Abstract
Microtubule actin cross-linking factor 1 (Macf1) is a spectraplakin family member known to regulate cytoskeletal dynamics, cell migration, neuronal growth and cell signal transduction. We previously demonstrated that knockdown of Macf1 inhibited the differentiation of MC3T3-E1 cell line. However, whether Macf1 could regulate bone formation in vivo is unclear. To study the function and mechanism of Macf1 in bone formation and osteogenic differentiation, we established osteoblast-specific Osterix (Osx) promoter-driven Macf1 conditional knockout mice (Macf1f/f Osx-Cre). The Macf1f/f Osx-Cre mice displayed delayed ossification and decreased bone mass. Morphological and mechanical studies showed deteriorated trabecular microarchitecture and impaired biomechanical strength of femur in Macf1f/f Osx-Cre mice. In addition, the differentiation of primary osteoblasts isolated from calvaria was inhibited in Macf1f/f Osx-Cre mice. Deficiency of Macf1 in primary osteoblasts inhibited the expression of osteogenic marker genes (Col1, Runx2 and Alp) and the number of mineralized nodules. Furthermore, deficiency of Macf1 attenuated Bmp2/Smad/Runx2 signalling in primary osteoblasts of Macf1f/f Osx-Cre mice. Together, these results indicated that Macf1 plays a significant role in bone formation and osteoblast differentiation by regulating Bmp2/Smad/Runx2 pathway, suggesting that Macf1 might be a therapeutic target for bone disease.
Collapse
Affiliation(s)
- Wu-Xia Qiu
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiao-Li Ma
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiao Lin
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Fan Zhao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Di-Jie Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Zhi-Hao Chen
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ke-Wen Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ru Zhang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Pai Wang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yun-Yun Xiao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Zhi-Ping Miao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Kai Dang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xiao-Yang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Ai-Rong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
21
|
Fenbo M, Xingyu X, Bin T. Strontium chondroitin sulfate/silk fibroin blend membrane containing microporous structure modulates macrophage responses for guided bone regeneration. Carbohydr Polym 2019; 213:266-275. [DOI: 10.1016/j.carbpol.2019.02.068] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 12/19/2022]
|
22
|
Lim ZXH, Rai B, Tan TC, Ramruttun AK, Hui JH, Nurcombe V, Teoh SH, Cool SM. Autologous bone marrow clot as an alternative to autograft for bone defect healing. Bone Joint Res 2019; 8:107-117. [PMID: 30997036 PMCID: PMC6444063 DOI: 10.1302/2046-3758.83.bjr-2018-0096.r1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objectives Long bone defects often require surgical intervention for functional restoration. The ‘gold standard’ treatment is autologous bone graft (ABG), usually from the patient’s iliac crest. However, autograft is plagued by complications including limited supply, donor site morbidity, and the need for an additional surgery. Thus, alternative therapies are being actively investigated. Autologous bone marrow (BM) is considered as a candidate due to the presence of both endogenous reparative cells and growth factors. We aimed to compare the therapeutic potentials of autologous bone marrow aspirate (BMA) and ABG, which has not previously been done. Methods We compared the efficacy of coagulated autologous BMA and ABG for the repair of ulnar defects in New Zealand White rabbits. Segmental defects (14 mm) were filled with autologous clotted BM or morcellized autograft, and healing was assessed four and 12 weeks postoperatively. Harvested ulnas were subjected to radiological, micro-CT, histological, and mechanical analyses. Results Comparable results were obtained with autologous BMA clot and ABG, except for the quantification of new bone by micro-CT. Significantly more bone was found in the ABG-treated ulnar defects than in those treated with autologous BMA clot. This is possibly due to the remnants of necrotic autograft fragments that persisted within the healing defects at week 12 post-surgery. Conclusion As similar treatment outcomes were achieved by the two strategies, the preferred treatment would be one that is associated with a lower risk of complications. Hence, these results demonstrate that coagulated BMA can be considered as an alternative autogenous therapy for long bone healing. Cite this article: Z. X. H. Lim, B. Rai, T. C. Tan, A. K. Ramruttun, J. H. Hui, V. Nurcombe, S. H. Teoh, S. M. Cool. Autologous bone marrow clot as an alternative to autograft for bone defect healing. Bone Joint Res 2019;8:107–117. DOI: 10.1302/2046-3758.83.BJR-2018-0096.R1.
Collapse
Affiliation(s)
- Z X H Lim
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - B Rai
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Science and Maths Cluster, Singapore University of Technology & Design (SUTD), Singapore
| | - T C Tan
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - A K Ramruttun
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - J H Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - V Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College, Singapore
| | - S H Teoh
- Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College, Singapore; School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - S M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
23
|
Wang X, Matthews BG, Yu J, Novak S, Grcevic D, Sanjay A, Kalajzic I. PDGF Modulates BMP2-Induced Osteogenesis in Periosteal Progenitor Cells. JBMR Plus 2019; 3:e10127. [PMID: 31131345 PMCID: PMC6524680 DOI: 10.1002/jbm4.10127] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/23/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
BMPs are used in various clinical applications to promote bone formation. The limited success of the BMPs in clinical settings and supraphysiological doses required for their effects prompted us to evaluate the influence of other signaling molecules, specifically platelet‐derived growth factor (PDGF) on BMP2‐induced osteogenesis. Periosteal cells make a major contribution to fracture healing. We detected broad expression of PDGF receptor beta (PDGFRβ) within the intact periosteum and healing callus during fracture repair. In vitro, periosteum‐derived progenitor cells were highly responsive to PDGF as demonstrated by increased proliferation and decreased apoptosis. However, PDGF blocked BMP2‐induced osteogenesis by inhibiting the canonical BMP2/Smad pathway and downstream target gene expression. This effect is mediated via PDGFRβ and involves ERK1/2 MAPK and PI3K/AKT signaling pathways. Therapeutic targeting of the PDGFRβ pathway in periosteum‐mediated bone repair might have profound implications in the treatment of bone disease in the future. © 2018 The Authors JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Xi Wang
- Department of Reconstructive Sciences UConn Health Farmington CT USA
| | - Brya G Matthews
- Department of Reconstructive Sciences UConn Health Farmington CT USA.,Department of Molecular Medicine and Pathology University of Auckland Auckland New Zealand
| | - Jungeun Yu
- Department of Orthopedic Surgery UConn Health Farmington CT USA
| | - Sanja Novak
- Department of Reconstructive Sciences UConn Health Farmington CT USA
| | - Danka Grcevic
- Department of Physiology and Immunology School of Medicine University of Zagreb Zagreb Croatia
| | - Archana Sanjay
- Department of Orthopedic Surgery UConn Health Farmington CT USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences UConn Health Farmington CT USA
| |
Collapse
|
24
|
Urbano N, Scimeca M, Bonanno E, Schillaci O. 99mTc sestamibi SPECT: a possible tool for early detection of breast cancer lesions with high bone metastatic potential. Future Oncol 2019; 15:455-457. [PMID: 30618271 DOI: 10.2217/fon-2018-0735] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Nicoletta Urbano
- Nuclear Medicine, Policlinico "Tor Vergata", viale Oxford, 81, 00133, Rome, Italy
| | - Manuel Scimeca
- Department of Biomedicine & Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.,IRCCS San Raffaele, Rome, Italy
| | - Elena Bonanno
- IRCCS San Raffaele, Rome, Italy.,Department of Experimental Medicine & Surgery, University of Rome "Tor Vergata", Via Montpellier, 1, 00133 Rome, Italy.,IRCCS Neuromed Lab. "Diagnostica Medica" & "Villa dei Platani", Avellino, Italy
| | - Orazio Schillaci
- Department of Biomedicine & Prevention, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
25
|
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
|
26
|
Conditional deletion of Bmp2 in cranial neural crest cells recapitulates Pierre Robin sequence in mice. Cell Tissue Res 2018; 376:199-210. [PMID: 30413887 DOI: 10.1007/s00441-018-2944-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/22/2018] [Indexed: 12/20/2022]
Abstract
Bone morphogenetic protein (BMP) signaling plays a crucial role in the development of craniofacial organs. Mutations in numerous members of the BMP signaling pathway lead to several severe human syndromes, including Pierre Robin sequence (PRS) caused by heterozygous loss of BMP2. In this study, we generate mice carrying Bmp2-specific deletion in cranial neural crest cells using floxed Bmp2 and Wnt1-Cre alleles to mimic PRS in humans. Mutant mice exhibit severe PRS with a significantly reduced size of craniofacial bones, cleft palate, malformed tongue and micrognathia. Palate clefting is caused by the undescended tongue that prevents palatal shelf elevation. However, the tongue in Wnt1-Cre;Bmp2f/f mice does not exhibit altered rates of cell proliferation and apoptosis, suggesting contribution of extrinsic defects to the failure of tongue descent. Further studies revealed obvious reduction in cell proliferation and differentiation of osteogenic progenitors in the mandible of the mutants, attributing to the micrognathia phenotype. Our study illustrates the pathogenesis of PRS caused by Bmp2 mutation, highlights the crucial role of BMP2 in the development of craniofacial bones and emphasizes precise coordination in the morphogenesis of palate, tongue and mandible during embryonic development.
Collapse
|
27
|
BMPRIA is required for osteogenic differentiation and RANKL expression in adult bone marrow mesenchymal stromal cells. Sci Rep 2018; 8:8475. [PMID: 29855498 PMCID: PMC5981611 DOI: 10.1038/s41598-018-26820-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/10/2018] [Indexed: 11/08/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) activate the canonical Smad1/5/8 and non-canonical Tak1-MAPK pathways via BMP receptors I and II to regulate skeletal development and bone remodeling. Specific ablation of Bmpr1a in immature osteoblasts, osteoblasts, or osteocytes results in an increase in cancellous bone mass, yet opposite results have been reported regarding the underlying mechanisms. Moreover, the role for BMPRIA-mediated signaling in bone marrow mesenchymal stromal cells (BM-MSCs) has not been explored. Here, we specifically ablated Bmpr1a in BM-MSCs in adult mice to study the function of BMPR1A in bone remodeling and found that the mutant mice showed an increase in cancellous and cortical bone mass, which was accompanied by a decrease in bone formation rate and a greater decrease in bone resorption. Decreased bone formation was associated with a defect in BM-MSC osteogenic differentiation whereas decreased bone resorption was associated with a decrease in RANKL production and osteoclastogenesis. However, ablation of Tak1, a critical non-canonical signaling molecule downstream of BMP receptors, in BM-MSCs at adult stage did not affect bone remodeling. These results suggest that BMP signaling through BMPRIA controls BM-MSC osteogenic differentiation/bone formation and RANKL expression/osteoclastogenesis in adult mice independent of Tak1 signaling.
Collapse
|
28
|
|
29
|
Grosso A, Burger MG, Lunger A, Schaefer DJ, Banfi A, Di Maggio N. It Takes Two to Tango: Coupling of Angiogenesis and Osteogenesis for Bone Regeneration. Front Bioeng Biotechnol 2017; 5:68. [PMID: 29164110 PMCID: PMC5675838 DOI: 10.3389/fbioe.2017.00068] [Citation(s) in RCA: 288] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 10/16/2017] [Indexed: 12/29/2022] Open
Abstract
Bone regeneration is a complex process requiring highly orchestrated interactions between different cells and signals to form new mineralized tissue. Blood vessels serve as a structural template, around which bone development takes place, and also bring together the key elements for bone homeostasis into the osteogenic microenvironment, including minerals, growth factors and osteogenic progenitor cells. Vascular endothelial growth factor (VEGF) is the master regulator of vascular growth and it is required for effective coupling of angiogenesis and osteogenesis during both skeletal development and postnatal bone repair. Here, we will review the current state of knowledge on the molecular cross-talk between angiogenesis and osteogenesis. In particular, we will focus on the role of VEGF in coupling these two processes and how VEGF dose can control the outcome, addressing in particular: (1) the direct influence of VEGF on osteogenic differentiation of mesenchymal progenitors; (2) the angiocrine functions of endothelium to regulate osteoprogenitors; (3) the role of immune cells, e.g., myeloid cells and osteoclast precursors, recruited by VEGF to the osteogenic microenvironment. Finally, we will discuss emerging strategies, based on the current biological understanding, to ensure rapid vascularization and efficient bone formation in regenerative medicine.
Collapse
Affiliation(s)
- Andrea Grosso
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| | - Maximilian G. Burger
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital of Basel, Basel, Switzerland
| | - Alexander Lunger
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital of Basel, Basel, Switzerland
| | - Dirk J. Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital of Basel, Basel, Switzerland
| | - Andrea Banfi
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| | - Nunzia Di Maggio
- Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| |
Collapse
|
30
|
Yanfeng T, Jianlin C, Yunbiao Z. [Experimental study on accelerated healing of jaw fracture using gelatin sponge compound growth factor]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2017; 35:506-509. [PMID: 29188647 DOI: 10.7518/hxkq.2017.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To explore the role and mechanism of drug delivery systems using growth factor combined with gelatin sponge on accelerating the healing of jaw fracture and to seek better treatment of accelerating the maxillofacial fracture. METHODS About 100 μg recombinant human bone morphogenetic protein (BMP)-2 was completely dissolved in 1 mL recombinant bovine basic fibroblast growth factor (bFGF), and the solution (40 μL) was dropped in gelatin sponge (0.5 cm×0.5 cm×1.0 cm). Then, it was freeze dried and prepared into bFGF/BMP/gelatin sponge delivery systems. The mandibular fracture model on two sides were prepared in 12 New Zealand rabbits and randomly divided into two groups. The left side was the control group, which was only fixed with titanium plates. The right side was the experimental group, in which bFGF/BMP/gelatin sponge delivery systems were put under the titanium plates. General observation, X-ray, and histological examination were taken at 2, 4, and 12 weeks after surgery. RESULTS After 2 weeks, more fibrous tissues were seen between the fracture ends in the experimental group than in the control group. After 4 weeks, fibrous fracture callus were seen in the fracture gap in the experimental group. The ingrowths of fibrous tissue and blood vessels were seen in the control group. The fracture healing of the experimental group was significantly faster than the control group at 2 and 4 weeks. After 12 weeks, the experimental and control groups all healed completely. CONCLUSIONS bFGF/BMP/gelatin sponge can accelerate and improve fracture healing; thus, it has better clinical application prospect.
Collapse
Affiliation(s)
- Tang Yanfeng
- Dept. of Oral and Maxillofacial Surgery, Baotou Central Hospital, Baotou 014040, China
| | - Chen Jianlin
- Dept. of Stomatology, Lingwu People's Hospital, Lingwu 750400, China
| | - Zhou Yunbiao
- Dept. of Oral and Maxillofacial Surgery, Baotou Central Hospital, Baotou 014040, China
| |
Collapse
|
31
|
BMP-2 induced Dspp transcription is mediated by Dlx3/Osx signaling pathway in odontoblasts. Sci Rep 2017; 7:10775. [PMID: 28883412 PMCID: PMC5589848 DOI: 10.1038/s41598-017-10908-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/16/2017] [Indexed: 01/18/2023] Open
Abstract
Dentin sialophosphoprotein (Dspp) as a differentiation marker of odontoblasts is regulated by BMP-2. However, the intimate mechanism is still unknown. Transcription factors Dlx3 and Osx are essential for odontoblasts differentiation. We hypothesized that BMP-2 regulation of Dspp transcription was mediated by Dlx3 and/or Osx in odontoblasts. In the present investigation, we found that BMP-2 stimulated expression and nuclear translocation of Dlx3 and Osx in odontoblasts both in vitro and in vivo. Osx was a downstream target of Dlx3 and both of them stimulated Dsp expression. Both Dlx3 and Osx were able to activate Dspp promoter from nucleotides (nt) -318 to +54 by transfections of luciferase reports containing different lengths of mouse Dspp promoters. The binding of Dlx3 and Osx with nt -318 to +54 of Dspp promoter was verified by chromatin immunoprecipitation in vivo. Two Dlx3 binding sites and one Osx binding site on Dspp promoter were found by EMSA. Furthermore, the exact biological function of these binding sites was confirmed by site-directed mutagenesis. At last, the protein-protein interaction between Dlx3 and Osx in odontoblasts was detected by co-immunoprecipitation. In conclusion, in this study we found a novel signaling pathway in which BMP-2 activates Dspp gene transcription via Dlx3/Osx pathway.
Collapse
|
32
|
BMP2 expression in the endocardial lineage is required for AV endocardial cushion maturation and remodeling. Dev Biol 2017; 430:113-128. [PMID: 28790014 DOI: 10.1016/j.ydbio.2017.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/16/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
Abstract
Distal outgrowth, maturation and remodeling of the endocardial cushion mesenchyme in the atrioventricular (AV) canal are the essential morphogenetic events during four-chambered heart formation. Mesenchymalized AV endocardial cushions give rise to the AV valves and the membranous ventricular septum (VS). Failure of these processes results in several human congenital heart defects. Despite this clinical relevance, the mechanisms governing how mesenchymalized AV endocardial cushions mature and remodel into the membranous VS and AV valves have only begun to be elucidated. The role of BMP signaling in the myocardial and secondary heart forming lineage has been well studied; however, little is known about the role of BMP2 expression in the endocardial lineage. To fill this knowledge gap, we generated Bmp2 endocardial lineage-specific conditional knockouts (referred to as Bmp2 cKOEndo) by crossing conditionally-targeted Bmp2flox/flox mice with a Cre-driver line, Nfatc1Cre, wherein Cre-mediated recombination was restricted to the endocardial cells and their mesenchymal progeny. Bmp2 cKOEndo mouse embryos did not exhibit failure or delay in the initial AV endocardial cushion formation at embryonic day (ED) 9.5-11.5; however, significant reductions in AV cushion size were detected in Bmp2 cKOEndo mouse embryos when compared to control embryos at ED13.5 and ED16.5. Moreover, deletion of Bmp2 from the endocardial lineage consistently resulted in membranous ventricular septal defects (VSDs), and mitral valve deficiencies, as evidenced by the absence of stratification of mitral valves at birth. Muscular VSDs were not found in Bmp2 cKOEndo mouse hearts. To understand the underlying morphogenetic mechanisms leading to a decrease in cushion size, cell proliferation and cell death were examined for AV endocardial cushions. Phospho-histone H3 analyses for cell proliferation and TUNEL assays for apoptotic cell death did not reveal significant differences between control and Bmp2 cKOEndo in AV endocardial cushions. However, mRNA expression of the extracellular matrix components, versican, Has2, collagen 9a1, and periostin was significantly reduced in Bmp2 cKOEndo AV cushions. Expression of transcription factors implicated in the cardiac valvulogenesis, Snail2, Twist1 and Sox9, was also significantly reduced in Bmp2 cKOEndo AV cushions. These data provide evidence that BMP2 expression in the endocardial lineage is essential for the distal outgrowth, maturation and remodeling of AV endocardial cushions into the normal membranous VS and the stratified AV valves.
Collapse
|
33
|
Hu K, Besschetnova TY, Olsen BR. Soluble VEGFR1 reverses BMP2 inhibition of intramembranous ossification during healing of cortical bone defects. J Orthop Res 2017; 35:1461-1469. [PMID: 27658810 DOI: 10.1002/jor.23416] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 09/06/2016] [Indexed: 02/04/2023]
Abstract
BMP2 is widely used for promotion of bone repair and regeneration. However, bone formation induced by BMP2 is quite variable. Bone forming progenitor cells in different locations appear to respond to BMP2 in different ways, and repair outcomes can vary as a consequence of modulating effects by other factors. In this study, we have examined the effects of VEGF on BMP2-induced repair of a cortical bone defect, a 1 mm diameter drill hole, in the proximal tibia of mice. Treatment of the defect with either a bolus of PBS or soluble VEGFR1 (sVEGFR1), a decoy receptor for VEGF, had the same effects on bone formation via intramembranous ossification in the defect and cartilage formation and injured periosteum, during the healing process. In contrast, treatment with BMP2 inhibited intramembranous bone formation in the defect while it promoted cartilage and endochondral bone formation in the injured periosteum compared with mice treated with PBS or sVEGFR1. The inhibitory effect of BMP2 on bone formation was unlikely due to increased osteoclast activity and decreased invasion of blood vessels in the defect. Most importantly, co-delivery of BMP2 and sVEGFR1 reversed the inhibition of intramembranous bone formation by BMP2. Furthermore, the decreased accumulation of collagen and production of bone matrix proteins in the defect of groups with BMP2 treatment could also be prevented by co-delivery of BMP2 and sVEGFR1. Our data indicate that introducing a VEGF-binding protein, such as sVEGFR1, to reduce levels of extracellular VEGF, may enhance the effects of BMP2 on intramembranous bone formation. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1461-1469, 2017.
Collapse
Affiliation(s)
- Kai Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Tatiana Y Besschetnova
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Shah TA, Zhu Y, Shaikh NN, Harris MA, Harris SE, Rogers MB. Characterization of new bone morphogenetic protein (Bmp)-2 regulatory alleles. Genesis 2017; 55. [PMID: 28401685 DOI: 10.1002/dvg.23035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/16/2017] [Accepted: 04/06/2017] [Indexed: 12/28/2022]
Abstract
Bone morphogenetic protein 2 (BMP2, HGNC:1069, GeneID: 650) is a classical morphogen; a molecule that acts at a distance and whose concentration influences cell proliferation, differentiation, and apoptosis. Key events requiring precise Bmp2 regulation include heart specification and morphogenesis and neural development. In mesenchymal cells, the concentration of BMP2 influences myogenesis, adipogenesis, chondrogenesis, and osteogenesis. Because the amount, timing, and location of BMP2 synthesis influence pattern formation and organogenesis, the mechanisms that regulate Bmp2 are crucial. A sequence within the 3'UTR of the Bmp2 mRNA termed the "ultra-conserved sequence" (UCS) has been largely unchanged since fishes and mammals diverged. Cre-lox mediated deletion of the UCS in a reporter transgene revealed that the UCS may repress Bmp2 in proepicardium, epicardium, and epicardium-derived cells (EPDC) and in tissues with known epicardial contributions (coronary vessels and valves). The UCS also repressed the transgene in the aorta, outlet septum, posterior cardiac plexus, cardiac and extra-cardiac nerves, and neural ganglia. We used homologous recombination and conditional deletion to generate three new alleles in which the Bmp2 3'UTR was altered as follows: a UCS flanked by loxP sites with or without a neomycin resistance targeting vector, or a deleted UCS. Deletion of the UCS was associated with elevated Bmp2 mRNA and BMP signaling levels, reduced fitness, and embryonic malformations.
Collapse
Affiliation(s)
- Tapan A Shah
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers NJMS, Newark, New Jersey
| | - Youhua Zhu
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers NJMS, Newark, New Jersey
| | - Nadia N Shaikh
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers NJMS, Newark, New Jersey
| | - Marie A Harris
- Department of Periodontics, University of Texas Health Science Centre, San Antonio, Texas
| | - Stephen E Harris
- Department of Periodontics, University of Texas Health Science Centre, San Antonio, Texas
| | - Melissa B Rogers
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers NJMS, Newark, New Jersey
| |
Collapse
|
35
|
Harris SE, Rediske M, Neitzke R, Rakian A. Periodontal Biology: Stem Cells, Bmp2 Gene, Transcriptional Enhancers, and Use of Sclerostin Antibody and Pth for Treatment of Periodontal Disease and Bone Loss. CELL, STEM CELLS AND REGENERATIVE MEDICINE 2017; 3:10.16966/2472-6990.113. [PMID: 29457146 PMCID: PMC5813290 DOI: 10.16966/2472-6990.113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The periodontium is a complex tissue with epithelial components and a complex set of mesodermal derived alveolar bone, cellular and a cellular cementum, and tendon like ligaments (PDL). The current evidence demonstrates that the major pool of periodontal stem cells is derived from a population of micro vascular associated aSMA-positive stem/progenitor (PSC) cells that by lineage tracing form all three major mesodermal derived components of the periodontium. With in vitro aSMA+ stem cells, transcriptome and chip- seq experiments, the gene network and enhancer maps were determined at several differentiation states of the PSC. Current work on the role of the Bmp2 gene in the periodontal stem cell differentiation demonstrated that this Wnt regulated gene, Bmp2, is necessary for differentiation to all three major mesodermal derived component of the periodontium. The mechanism and use of Sclerostin antibody as an activator of Wnt signaling and Bmp2 gene as a potential route to treat craniofacial bone loss is discussed. As well, the mechanism and use of Pth in the treatment of periodontal bone loss or other craniofacial bone loss is presented in this review.
Collapse
Affiliation(s)
- Stephen E Harris
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Michael Rediske
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Rebecca Neitzke
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Audrey Rakian
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
36
|
Wang T, Zhang X, Bikle DD. Osteogenic Differentiation of Periosteal Cells During Fracture Healing. J Cell Physiol 2016; 232:913-921. [PMID: 27731505 DOI: 10.1002/jcp.25641] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/07/2016] [Indexed: 12/15/2022]
Abstract
Five to ten percent of fractures fail to heal normally leading to additional surgery, morbidity, and altered quality of life. Fracture healing involves the coordinated action of stem cells primarily coming from the periosteum which differentiate into the chondrocytes and osteoblasts, forming first the soft (cartilage) callus followed by the hard (bone) callus. These stem cells are accompanied by a vascular invasion that appears critical for the differentiation process and which may enable the entry of osteoclasts necessary for the remodeling of the callus into mature bone. However, more research is needed to clarify the signaling events that activate the osteochondroprogenitor cells of periosteum and stimulate their differentiation into chondrocytes and osteoblasts. Ultimately a thorough understanding of the mechanisms for differential regulation of these osteochondroprogenitors will aid in the treatment of bone healing and the prevention of delayed union and nonunion of fractures. In this review, evidence supporting the concept that the periosteal cells are the major cell sources of skeletal progenitors for the fracture callus will be discussed. The osteogenic differentiation of periosteal cells manipulated by Wnt/β-catenin, TGF/BMP, Ihh/PTHrP, and IGF-1/PI3K-Akt signaling in fracture repair will be examined. The effect of physical (hypoxia and hyperoxia) and chemical factors (reactive oxygen species) as well as the potential coordinated regulatory mechanisms in the periosteal progenitor cells promoting osteogenic differentiation will also be discussed. Understanding the regulation of periosteal osteochondroprogenitors during fracture healing could provide insight into possible therapeutic targets and thereby help to enhance future fracture healing and bone tissue engineering approaches. J. Cell. Physiol. 232: 913-921, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tao Wang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York.,Endocrine Unit, VA Medical Center and University of California, San Francisco, California
| | - Xinping Zhang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Daniel D Bikle
- Endocrine Unit, VA Medical Center and University of California, San Francisco, California
| |
Collapse
|
37
|
Salazar VS, Ohte S, Capelo LP, Gamer L, Rosen V. Specification of osteoblast cell fate by canonical Wnt signaling requires Bmp2. Development 2016; 143:4352-4367. [PMID: 27802170 DOI: 10.1242/dev.136879] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 10/07/2016] [Indexed: 12/14/2022]
Abstract
Enhanced BMP or canonical Wnt (cWnt) signaling are therapeutic strategies employed to enhance bone formation and fracture repair, but the mechanisms each pathway utilizes to specify cell fate of bone-forming osteoblasts remain poorly understood. Among all BMPs expressed in bone, we find that singular deficiency of Bmp2 blocks the ability of cWnt signaling to specify osteoblasts from limb bud or bone marrow progenitors. When exposed to cWnts, Bmp2-deficient cells fail to progress through the Runx2/Osx1 checkpoint and thus do not upregulate multiple genes controlling mineral metabolism in osteoblasts. Cells lacking Bmp2 after induction of Osx1 differentiate normally in response to cWnts, suggesting that pre-Osx1+ osteoprogenitors are an essential source and a target of BMP2. Our analysis furthermore reveals Grainyhead-like 3 (Grhl3) as a transcription factor in the osteoblast gene regulatory network induced during bone development and bone repair, which acts upstream of Osx1 in a BMP2-dependent manner. The Runx2/Osx1 transition therefore receives crucial regulatory inputs from BMP2 that are not compensated for by cWnt signaling, and this is mediated at least in part by induction and activation of Grhl3.
Collapse
Affiliation(s)
- Valerie S Salazar
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| | - Satoshi Ohte
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA.,Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Luciane P Capelo
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA.,Instituto de Ciência e Tecnologia, Universidade Federal de São Paulo, Rua Talim, 330, São José dos Campos, São Paulo, CEP 12231-280, Brazil
| | - Laura Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
38
|
Ozeki N, Mogi M, Hase N, Hiyama T, Yamaguchi H, Kawai R, Matsumoto T, Nakata K. Bone morphogenetic protein-induced cell differentiation involves Atg7 and Wnt16 sequentially in human stem cell-derived osteoblastic cells. Exp Cell Res 2016; 347:24-41. [PMID: 27397580 DOI: 10.1016/j.yexcr.2016.07.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023]
Abstract
We established a differentiation method for homogeneous α7 integrin-positive human skeletal muscle stem cell (α7(+)hSMSC)-derived osteoblast-like cells with bone morphogenetic protein (BMP)-2. To explore the early signaling cascade for osteoblastic differentiation, we examined the upregulation of autophagy-related gene (Atg) and wingless/int1 (Wnt) signaling during BMP-2-mediated human osteoblastic differentiation. In a screening experiment, BMP-2 increased the mRNA and protein levels of Atg7, Wnt16, and Lrp5/Fzd2 (a Wnt receptor), but not microtubule-associated protein 1 light chain (LC3; a mammalian homolog of yeast Atg8), TFE3, Beclin1, Atg5, Atg12, Wnt3a, or Wnt5, together with the amounts of autophagosomes and autophagy fluxes. Treatment with siRNAs against Atg7 and Wnt16 individually suppressed the BMP-2-induced increase in osteoblastic differentiation. The osteoblastic phenotype, involving osteocalcin (BGLAP), osteopontin (SPP1), and osterix (SP7) expression, decreased when autophagy was inhibited by chloroquine (an autophagy inhibitor), but increased after treatment with rapamycin (an autophagy enhancer). Taken together with our previous findings, we have revealed a unique sequential cascade of BMP-2→Atg7→Wnt16→Lrp5/Fzd2→matrix metalloproteinase-13→osteoblastic differentiation. This cascade results in a potent increase in osteoblastic cell differentiation, indicating the unique involvement of Atg7, autophagy, and Wnt16 signaling in BMP-2-induced differentiation of α7(+)hSMSCs into osteoblast-like cells at a relatively early stage.
Collapse
Affiliation(s)
- Nobuaki Ozeki
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi 464-8651, Japan.
| | - Makio Mogi
- Department of Integrative Education of Pharmacy, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Naoko Hase
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi 464-8651, Japan
| | - Taiki Hiyama
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi 464-8651, Japan
| | - Hideyuki Yamaguchi
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi 464-8651, Japan
| | - Rie Kawai
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi 464-8651, Japan
| | - Toru Matsumoto
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi 464-8651, Japan
| | - Kazuhiko Nakata
- Department of Endodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, Aichi 464-8651, Japan
| |
Collapse
|
39
|
Hydrogel Delivery of Mesenchymal Stem Cell-Expressing Bone Morphogenetic Protein-2 Enhances Bone Defect Repair. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2016; 4:e838. [PMID: 27622106 PMCID: PMC5010329 DOI: 10.1097/gox.0000000000000817] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 05/17/2016] [Indexed: 01/24/2023]
Abstract
BACKGROUND The application of bone tissue engineering for repairing bone defects has gradually shown some satisfactory progress. One of the concerns raising scientific attention is the poor supply of growth factors. A number of growth factor delivery approaches have been developed for promoting bone formation. However, there is no systematic comparison of those approaches on efficiency of neobone formation. In this study, the approaches using periosteum, direct supply of growth factors, or gene transfection of growth factors were evaluated to determine the osteogenic capacity on the repair of bone defect. METHODS In total, 42 male 21-week-old Sprague-Dawley rats weighing 250 to 400 g were used as the bone defect model to evaluate the bone repair efficiency. Various tissue engineered constructs of poly(ethylene glycol)-poly(l-lactic acid) (PEG-PLLA) copolymer hydrogel with periosteum, with external supply of bone morphogenetic protein-2 (BMP2), or with BMP2-transfected bone marrow-derived mesenchymal stem cells (BMMSCs) were filled in a 7-mm bone defect region. Animals were euthanized at 3 months, and the hydrogel constructs were harvested. The evaluation with histological staining and radiography analysis were performed for the volume of new bone formation. RESULTS The PEG-PLLA scaffold with BMMSCs promotes bone regeneration with the addition of periosteum. The group with BMP2-transfected BMMSCs demonstrated the largest volume of new bone among all the testing groups. CONCLUSIONS Altogether, the results of this study provide the evidence that the combination of PEG-PLLA hydrogels with BMMSCs and sustained delivery of BMP2 resulted in the maximal bone regeneration.
Collapse
|
40
|
Zhang Y, McNerny EG, Terajima M, Raghavan M, Romanowicz G, Zhang Z, Zhang H, Kamiya N, Tantillo M, Zhu P, Scott GJ, Ray MK, Lynch M, Ma PX, Morris MD, Yamauchi M, Kohn DH, Mishina Y. Loss of BMP signaling through BMPR1A in osteoblasts leads to greater collagen cross-link maturation and material-level mechanical properties in mouse femoral trabecular compartments. Bone 2016; 88:74-84. [PMID: 27113526 PMCID: PMC4899267 DOI: 10.1016/j.bone.2016.04.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 03/26/2016] [Accepted: 04/22/2016] [Indexed: 01/23/2023]
Abstract
Bone morphogenetic protein (BMP) signaling pathways play critical roles in skeletal development and new bone formation. Our previous study, however, showed a negative impact of BMP signaling on bone mass because of the osteoblast-specific loss of a BMP receptor (i.e. BMPR1A) showing increased trabecular bone volume and mineral density in mice. Here, we investigated the bone quality and biomechanical properties of the higher bone mass associated with BMPR1A deficiency using the osteoblast-specific Bmpr1a conditional knockout (cKO) mouse model. Collagen biochemical analysis revealed greater levels of the mature cross-link pyridinoline in the cKO bones, in parallel with upregulation of collagen modifying enzymes. Raman spectroscopy distinguished increases in the mature to immature cross-link ratio and mineral to matrix ratio in the trabecular compartments of cKO femora, but not in the cortical compartments. The mineral crystallinity was unchanged in the cKO in either the trabecular or cortical compartments. Further, we tested the intrinsic material properties by nanoindentation and found significantly higher hardness and elastic modulus in the cKO trabecular compartments, but not in the cortical compartments. Four point bending tests of cortical compartments showed lower structural biomechanical properties (i.e. strength and stiffness) in the cKO bones due to the smaller cortical areas. However, there were no significant differences in biomechanical performance at the material level, which was consistent with the nanoindentation test results on the cortical compartment. These studies emphasize the pivotal role of BMPR1A in the determination of bone quality and mechanical integrity under physiological conditions, with different impact on femoral cortical and trabecular compartments.
Collapse
Affiliation(s)
- 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
| | - Mekhala Raghavan
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Genevieve Romanowicz
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Zhanpeng Zhang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Honghao Zhang
- 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; Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219, USA; Faculty of Budo and Sport Studies, Tenri University, Nara, Japan
| | - Margaret Tantillo
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - Peizhi Zhu
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan, MI, USA
| | - Gregory J Scott
- Knock Out Core, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Manas K Ray
- Knock Out Core, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Michelle Lynch
- Office of Research, School of Dentistry, University of Michigan, MI, USA
| | - Peter X Ma
- Department of Biologic and Materials Sciences, School of Dentistry, 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; Biomedical Engineering, College of Engineering, University of Michigan, MI, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA; Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; Knock Out Core, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
41
|
Regenerative Engineering in Maxillofacial Reconstruction. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2016. [DOI: 10.1007/s40883-016-0009-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
42
|
Abstract
Since the identification in 1988 of bone morphogenetic protein 2 (BMP2) as a potent inducer of bone and cartilage formation, BMP superfamily signalling has become one of the most heavily investigated topics in vertebrate skeletal biology. Whereas a large part of this research has focused on the roles of BMP2, BMP4 and BMP7 in the formation and repair of endochondral bone, a large number of BMP superfamily molecules have now been implicated in almost all aspects of bone, cartilage and joint biology. As modulating BMP signalling is currently a major therapeutic target, our rapidly expanding knowledge of how BMP superfamily signalling affects most tissue types of the skeletal system creates enormous potential to translate basic research findings into successful clinical therapies that improve bone mass or quality, ameliorate diseases of skeletal overgrowth, and repair damage to bone and joints. This Review examines the genetic evidence implicating BMP superfamily signalling in vertebrate bone and joint development, discusses a selection of human skeletal disorders associated with altered BMP signalling and summarizes the status of modulating the BMP pathway as a therapeutic target for skeletal trauma and disease.
Collapse
Affiliation(s)
- Valerie S Salazar
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Laura W Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
43
|
Ren L, Chen X, Ouyang H. Interactions of porcine circovirus 2 with its hosts. Virus Genes 2016; 52:437-44. [DOI: 10.1007/s11262-016-1326-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/19/2016] [Indexed: 12/11/2022]
|
44
|
McBride-Gagyi SH, McKenzie JA, Buettmann EG, Gardner MJ, Silva MJ. Bmp2 conditional knockout in osteoblasts and endothelial cells does not impair bone formation after injury or mechanical loading in adult mice. Bone 2015; 81:533-543. [PMID: 26344756 PMCID: PMC4640950 DOI: 10.1016/j.bone.2015.09.003] [Citation(s) in RCA: 34] [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] [Received: 06/04/2015] [Revised: 08/20/2015] [Accepted: 09/03/2015] [Indexed: 12/11/2022]
Abstract
Post-natal osteogenesis after mechanical trauma or stimulus occurs through either endochondral healing, intramembranous healing or lamellar bone formation. Bone morphogenetic protein 2 (BMP2) is up-regulated in each of these osteogenic processes and is expressed by a variety of cells including osteoblasts and vascular cells. It is known that genetic knockout of Bmp2 in all cells or in osteo-chondroprogenitor cells completely abrogates endochondral healing after full fracture. However, the importance of BMP2 from differentiated osteoblasts and endothelial cells is not known. Moreover, the importance of BMP2 in non-endochondral bone formation such as intramembranous healing or lamellar bone formation is not known. Using inducible and tissue-specific Cre-lox mediated targeting of Bmp2 in adult (10-24 week old) mice, we assessed the role of BMP2 expression globally, by osteoblasts, and by vascular endothelial cells in endochondral healing, intramembranous healing and lamellar bone formation. These three osteogenic processes were modeled using full femur fracture, ulnar stress fracture, and ulnar non-damaging cyclic loading, respectively. Our results confirmed the requirement of BMP2 for endochondral fracture healing, as mice in which Bmp2 was knocked out in all cells prior to fracture failed to form a callus. Targeted deletion of Bmp2 in osteoblasts (osterix-expressing) or vascular endothelial cells (vascular endothelial cadherin-expressing) did not impact fracture healing in any way. Regarding non-endochondral bone formation, we found that BMP2 is largely dispensable for intramembranous bone formation after stress fracture and also not required for lamellar bone formation induced by mechanical loading. Taken together our results indicate that osteoblasts and endothelial cells are not a critical source of BMP2 in endochondral fracture healing, and that non-endochondral bone formation in the adult mouse is not as critically dependent on BMP2.
Collapse
Affiliation(s)
- Sarah Howe McBride-Gagyi
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall, M176, St. Louis, MO 63104, USA; Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Jennifer A McKenzie
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Evan G Buettmann
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Michael J Gardner
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Matthew J Silva
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| |
Collapse
|
45
|
Mandal CC, Das F, Ganapathy S, Harris SE, Choudhury GG, Ghosh-Choudhury N. Bone Morphogenetic Protein-2 (BMP-2) Activates NFATc1 Transcription Factor via an Autoregulatory Loop Involving Smad/Akt/Ca2+ Signaling. J Biol Chem 2015; 291:1148-61. [PMID: 26472929 DOI: 10.1074/jbc.m115.668939] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Indexed: 11/06/2022] Open
Abstract
Bone remodeling is controlled by dual actions of osteoclasts (OCs) and osteoblasts (OBs). The calcium-sensitive nuclear factor of activated T cells (NFAT) c1 transcription factor, as an OC signature gene, regulates differentiation of OCs downstream of bone morphogenetic protein-2 (BMP-2)-stimulated osteoblast-coded factors. To analyze a functional link between BMP-2 and NFATc1, we analyzed bones from OB-specific BMP-2 knock-out mice for NFATc1 expression by immunohistochemical staining and found significant reduction in NFATc1 expression. This indicated a requirement of BMP-2 for NFATc1 expression in OBs. We showed that BMP-2, via the receptor-specific Smad pathway, regulates expression of NFATc1 in OBs. Phosphatidylinositol 3-kinase/Akt signaling acting downstream of BMP-2 also drives NFATc1 expression and transcriptional activation. Under the basal condition, NFATc1 is phosphorylated. Activation of NFAT requires dephosphorylation by the calcium-dependent serine/threonine phosphatase calcineurin. We examined the role of calcium in BMP-2-stimulated regulation of NFATc1 in osteoblasts. 1,2Bis(2aminophenoxy)ethaneN,N,N',N'-tetraacetic acid acetoxymethyl ester, an inhibitor of intracellular calcium abundance, blocked BMP-2-induced transcription of NFATc1. Interestingly, BMP-2 induced calcium release from intracellular stores and increased calcineurin phosphatase activity, resulting in NFATc1 nuclear translocation. Cyclosporin A, which inhibits calcineurin upstream of NFATc1, blocked BMP-2-induced NFATc1 mRNA and protein expression. Expression of NFATc1 directly increased its transcription and VIVIT peptide, an inhibitor of NFATc1, suppressed BMP-2-stimulated NFATc1 transcription, confirming its autoregulation. Together, these data show a role of NFATc1 downstream of BMP-2 in mouse bone development and provide novel evidence for the presence of a cross-talk among Smad, phosphatidylinositol 3-kinase/Akt, and Ca(2+) signaling for BMP-2-induced NFATc1 expression through an autoregulatory loop.
Collapse
Affiliation(s)
| | | | | | - Stephen E Harris
- Periodontics, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Goutam Ghosh Choudhury
- Medicine, and From Veterans Affairs Research and Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System and
| | | |
Collapse
|
46
|
Santos L, Fuhrmann G, Juenet M, Amdursky N, Horejs CM, Campagnolo P, Stevens MM. Extracellular Stiffness Modulates the Expression of Functional Proteins and Growth Factors in Endothelial Cells. Adv Healthc Mater 2015; 4:2056-2063. [PMID: 26270789 DOI: 10.1002/adhm.201500338] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/25/2015] [Indexed: 01/08/2023]
Abstract
Angiogenesis, the formation of blood vessels from pre-existing ones, is of vital importance during the early stages of bone healing. Extracellular stiffness plays an important role in regulating endothelial cell behavior and angiogenesis, but how this mechanical cue affects proliferation kinetics, gene regulation, and the expression of proteins implicated in angiogenesis and bone regeneration remains unclear. Using collagen-coated polyacrylamide (PAAm) hydrogels, human umbilical vein endothelial cells (HUVECs) are exposed to an environment that mimics the elastic properties of collagenous bone, and cellular proliferation and gene and protein expressions are assessed. The proliferation and gene expression of HUVECs are not differentially affected by culture on 3 or 30 kPa PAAm hydrogels, henceforth referred to as low and high stiffness gels, respectively. Although the proliferation and gene transcript levels remain unchanged, significant differences are found in the expressions of functional proteins and growth factors implicated both in the angiogenic and osteogenic processes. The down-regulation of the vascular endothelial growth factor receptor-2 protein with concomitant over-expression of caveolin-1, wingless-type 2, bone morphogenic protein 2, and basic fibroblast growth factor on the high stiffness PAAm hydrogel suggests that rigidity has a pro-angiogenic effect with inherent benefits for bone regeneration.
Collapse
Affiliation(s)
- Lívia Santos
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Gregor Fuhrmann
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Maya Juenet
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Nadav Amdursky
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Christine-Maria Horejs
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Paola Campagnolo
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Molly M. Stevens
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| |
Collapse
|
47
|
Guo F, Feng J, Wang F, Li W, Gao Q, Chen Z, Shoff L, Donly KJ, Gluhak-Heinrich J, Chun YHP, Harris SE, MacDougall M, Chen S. Bmp2 deletion causes an amelogenesis imperfecta phenotype via regulating enamel gene expression. J Cell Physiol 2015; 230:1871-82. [PMID: 25545831 DOI: 10.1002/jcp.24915] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 12/09/2014] [Accepted: 12/18/2014] [Indexed: 01/16/2023]
Abstract
Although Bmp2 is essential for tooth formation, the role of Bmp2 during enamel formation remains unknown in vivo. In this study, the role of Bmp2 in regulation of enamel formation was investigated by the Bmp2 conditional knock out (Bmp2 cKO) mice. Teeth of Bmp2 cKO mice displayed severe and profound phenotypes with asymmetric and misshaped incisors as well as abrasion of incisors and molars. Scanning electron microscopy analysis showed that the enamel layer was hypoplastic and enamel lacked a typical prismatic pattern. Teeth from null mice were much more brittle as tested by shear and compressive moduli. Expression of enamel matrix protein genes, amelogenin, enamelin, and enamel-processing proteases, Mmp-20 and Klk4 was reduced in the Bmp2 cKO teeth as reflected in a reduced enamel formation. Exogenous Bmp2 up-regulated those gene expressions in mouse enamel organ epithelial cells. This result for the first time indicates Bmp2 signaling is essential for proper enamel development and mineralization in vivo.
Collapse
Affiliation(s)
- Feng Guo
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Department of Developmental Dentistry, The University of Texas Health Science Center at San Antonio, Floyd Curl Drive, San Antonio, Texas
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Intini G, Nyman JS. Dkk1 haploinsufficiency requires expression of Bmp2 for bone anabolic activity. Bone 2015; 75:151-60. [PMID: 25603465 PMCID: PMC4387090 DOI: 10.1016/j.bone.2015.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 12/09/2014] [Accepted: 01/13/2015] [Indexed: 12/17/2022]
Abstract
Bone fractures remain a serious health burden and prevention and enhanced healing of fractures have been obtained by augmenting either BMP or Wnt signaling. However, whether BMP and Wnt signaling are both required or are self-sufficient for anabolic and fracture healing activities has never been fully elucidated. Mice haploinsufficient for Dkk1 (Dkk1(+/-)) exhibit a high bone mass phenotype due to an up-regulation of canonical Wnt signaling while mice lacking Bmp2 expression in the limbs (Bmp2(c/c);Prx1::cre) succumb to spontaneous fracture and are unable to initiate fracture healing; combined, these mice offer an opportunity to examine the requirement for activated BMP signaling on the anabolic and fracture healing activity of Wnts. When Dkk1(+/-) mice were crossed with Bmp2(c/c);Prx1::cre mice, the offspring bearing both genetic alterations were unable to increase bone mass and heal fractures, indicating that increased canonical Wnt signaling is unable to exploit its activity in absence of Bmp2. Thus, our data suggest that BMP signaling is required for Wnt-mediated anabolic activity and that therapies aimed at preventing fractures and fostering fracture repair may need to target both pathways for maximal efficacy.
Collapse
Affiliation(s)
- Giuseppe Intini
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, 185 Cambridge Street, Boston, MA 02114, USA.
| | - Jeffry S Nyman
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
49
|
Agas D, Marchetti L, Douni E, Sabbieti MG. The unbearable lightness of bone marrow homeostasis. Cytokine Growth Factor Rev 2015; 26:347-59. [DOI: 10.1016/j.cytogfr.2014.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/22/2014] [Accepted: 12/17/2014] [Indexed: 01/10/2023]
|
50
|
Lipocalin produced by myelofibrosis cells affects the fate of both hematopoietic and marrow microenvironmental cells. Blood 2015; 126:972-82. [PMID: 26022238 DOI: 10.1182/blood-2014-12-618595] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/18/2015] [Indexed: 12/20/2022] Open
Abstract
Myelofibrosis (MF) is characterized by cytopenias, constitutional symptoms, splenomegaly, and marrow histopathological abnormalities (fibrosis, increased microvessel density, and osteosclerosis). The microenvironmental abnormalities are likely a consequence of the elaboration of a variety of inflammatory cytokines generated by malignant megakaryocytes and monocytes. We observed that levels of a specific inflammatory cytokine, lipocalin-2 (LCN2), were elevated in the plasmas of patients with myeloproliferative neoplasms (MF > polycythemia vera or essential thrombocythemia) and that LCN2 was elaborated by MF myeloid cells. LCN2 generates increased reactive oxygen species, leading to increased DNA strand breaks and apoptosis of normal, but not MF, CD34(+) cells. Furthermore, incubation of marrow adherent cells or mesenchymal stem cells with LCN2 increased the generation of osteoblasts and fibroblasts, but not adipocytes. LCN2 priming of mesenchymal stem cells resulted in the upregulation of RUNX2 gene as well as other genes that are capable of further affecting osteoblastogenesis, angiogenesis, and the deposition of matrix proteins. These data indicate that LCN2 is an additional MF inflammatory cytokine that likely contributes to the creation of a cascade of events that results in not only a predominance of the MF clone but also a dysfunctional microenvironment.
Collapse
|