1
|
Almalki MH, Almohaya M, Aziz F, AlDahmani KM, Alashgar L, Ekhzaimy A, Mahzari M. Prevalence of vertebral fractures in patients with acromegaly: A multicenter cross-sectional study. Hormones (Athens) 2025:10.1007/s42000-025-00652-4. [PMID: 40210828 DOI: 10.1007/s42000-025-00652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
INTRODUCTION Acromegaly is a chronic disease characterized by excessive growth hormone (GH) secretion leading to high insulin-like growth factor 1 (IGF- 1) levels. It results in systemic complications, including vertebral fractures (VFs), which impair quality of life. Bone mineral density (BMD) may not fully capture fracture risk, warranting further research into alternative predictors and mitigation strategies. OBJECTIVES This study aimed to assess the prevalence of VFs in patients with acromegaly and identify associated risk factors. METHODS A multicenter, cross-sectional study was conducted at three centers in Saudi Arabia and one in the United Arab Emirates (UAE). Data for patients with acromegaly evaluated between 2010 and 2024 were reviewed. RESULTS The cohort consisted of 101 patients (63% male) with a median age of 45 years and an median disease duration of 7 years. VFs were present in 10 patients, predominantly affecting the lumbar spine and were generally mild. Although not statistically significant, patients with VFs had longer disease durations. BMD measurements did not significantly differ between those with and without VFs; however, patients with VFs showed a trend toward lower trabecular bone score (TBS), suggesting compromised bone microarchitecture. CONCLUSION This study underscores the importance of comprehensive bone health assessment in patients with acromegaly incorporating BMD, TBS, and hormonal evaluations. Continuous long-term monitoring of bone health parameters is necessary in this population. Future research is needed to delineate risk factors associated with VFs.
Collapse
Affiliation(s)
- Mussa H Almalki
- Obesity, Endocrine, and Metabolism Center, King Fahad Medical City, Second Health Cluster, Riyadh, Saudi Arabia.
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| | - Mohammed Almohaya
- Obesity, Endocrine, and Metabolism Center, King Fahad Medical City, Second Health Cluster, Riyadh, Saudi Arabia
| | - Faisal Aziz
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Khaled M AlDahmani
- Division of Endocrinology, Department of Medicine, Tawam Hospital, Al Ain, United Arab Emirates
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Division of Endocrinology, Department of Medicine, Sheikh Tahnoon Bin Mohammed Medical City, Al Ain, United Arab Emirates
| | - Lolwah Alashgar
- Endocrinology and Diabetes Unit, Department of Medicine, College of Medicine and King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Aishah Ekhzaimy
- Endocrinology and Diabetes Unit, Department of Medicine, College of Medicine and King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Moeber Mahzari
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Medicine, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Berger DR, Centeno CJ, Kisiday JD, McIlwraith CW, Steinmetz NJ. Colony Forming Potential and Protein Composition of Commercial Umbilical Cord Allograft Products in Comparison With Autologous Orthobiologics. Am J Sports Med 2021; 49:3404-3413. [PMID: 34398643 DOI: 10.1177/03635465211031275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Umbilical cord (UC) connective tissues contain plastic-adherent, colony forming unit-fibroblasts (CFU-Fs) amenable to culture expansion for potential therapeutic use. Recently, UC-derived allograft products have been made available to practitioners in orthopaedics and other specialties, by companies purporting "stem cell"-based healing. However, such marketing claims conflict with existing regulations for these human tissues, generating questions over the cellular and protein composition of current commercially available UC allograft products. PURPOSE To evaluate commercial UC allograft products for viable cells, CFU-Fs, and protein makeup. STUDY DESIGN Descriptive laboratory study. METHODS Five commercial UC allograft products claiming to contain viable, undescribed "stem cells," 2 obtained from UC blood (UCB) and 3 from UC tissue (UCT), were analyzed. Image-based methods were used to measure cell concentration and viability, a traditional CFU-F assay was used to evaluate in vitro behavior indicative of a connective tissue progenitor cell phenotype often referred to as mesenchymal stem/stromal cells, and quantitative immunoassay arrays were used to measure a combination of cytokines and growth factors. Bone marrow concentrate (BMC) and plasma derived from the blood and bone marrow of middle-aged individuals served as comparative controls for cell culture and protein analyses, respectively. RESULTS Viable cells were identified within all 5 UC allograft products, with those derived from UCB having greater percentages of living cells (40%-59%) than those from UCT (1%-22%). Compared with autologous BMC (>95% viability and >300 million living cells), no CFU-Fs were observed within any UC allograft product (<15 million living cells). Moreover, a substantial number of proteins, particularly those within UCB allograft products, were undetectable or present at lower concentrations compared with blood and bone marrow plasma controls. Interestingly, several important growth factors and cytokines, including basic fibroblast growth factor, hepatocyte growth factor, interleukin-1 receptor antagonist, and osteoprotegerin, were most prevalent in 1 or more UCT allograft products as compared with blood and bone marrow plasma. CONCLUSION CFU-Fs, often referred to as stem cells, were not found within any of the commercial UC allograft products analyzed, and clinicians should remain wary of marketing claims stating otherwise. CLINICAL RELEVANCE Any therapeutic benefit of current UC allograft products in orthopaedic medicine is more likely to be attributed to their protein composition (UCT > UCB) or inclusion of cells without colony forming potential (UCB > UCT).
Collapse
Affiliation(s)
- Dustin R Berger
- Research and Development, Regenexx, LLC, Broomfield, Colorado, USA
| | - Christopher J Centeno
- Research and Development, Regenexx, LLC, Broomfield, Colorado, USA
- Centeno-Schultz Clinic, Broomfield, Colorado, USA
| | - John D Kisiday
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - C Wayne McIlwraith
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Science, Colorado State University, Fort Collins, Colorado, USA
| | | |
Collapse
|
3
|
Tonomura H, Nagae M, Takatori R, Ishibashi H, Itsuji T, Takahashi K. The Potential Role of Hepatocyte Growth Factor in Degenerative Disorders of the Synovial Joint and Spine. Int J Mol Sci 2020; 21:ijms21228717. [PMID: 33218127 PMCID: PMC7698933 DOI: 10.3390/ijms21228717] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/30/2020] [Accepted: 11/16/2020] [Indexed: 02/08/2023] Open
Abstract
This paper aims to provide a comprehensive review of the changing role of hepatocyte growth factor (HGF) signaling in the healthy and diseased synovial joint and spine. HGF is a multifunctional growth factor that, like its specific receptor c-Met, is widely expressed in several bone and joint tissues. HGF has profound effects on cell survival and proliferation, matrix metabolism, inflammatory response, and neurotrophic action. HGF plays an important role in normal bone and cartilage turnover. Changes in HGF/c-Met have also been linked to pathophysiological changes in degenerative joint diseases, such as osteoarthritis (OA) and intervertebral disc degeneration (IDD). A therapeutic role of HGF has been proposed in the regeneration of osteoarticular tissues. HGF also influences bone remodeling and peripheral nerve activity. Studies aimed at elucidating the changing role of HGF/c-Met signaling in OA and IDD at different pathophysiological stages, and their specific molecular mechanisms are needed. Such studies will contribute to safe and effective HGF/c-Met signaling-based treatments for OA and IDD.
Collapse
|
4
|
Sun Z, Cai S, Zabkiewicz C, Liu C, Ye L. Bone morphogenetic proteins mediate crosstalk between cancer cells and the tumour microenvironment at primary tumours and metastases (Review). Int J Oncol 2020; 56:1335-1351. [PMID: 32236571 DOI: 10.3892/ijo.2020.5030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/28/2020] [Indexed: 11/05/2022] Open
Abstract
Bone morphogenetic proteins (BMP) are pluripotent molecules, co‑ordinating cellular functions from early embryonic and postnatal development to tissue repair, regeneration and homeostasis. They are also involved in tumourigenesis, disease progression and the metastasis of various solid tumours. Emerging evidence has indicated that BMPs are able to promote disease progression and metastasis by orchestrating communication between cancer cells and the surrounding microenvironment. The interactions occur between BMPs and epidermal growth factor receptor, hepatocyte growth factor, fibroblast growth factor, vascular endothelial growth factor and extracellular matrix components. Overall, these interactions co‑ordinate the cellular functions of tumour cells and other types of cell in the tumour to promote the growth of the primary tumour, local invasion, angiogenesis and metastasis, and the establishment and survival of cancer cells in the metastatic niche. Therefore, the present study aimed to provide an informative summary of the involvement of BMPs in the tumour microenvironment.
Collapse
Affiliation(s)
- Zhiwei Sun
- VIP‑II Division of Medical Department, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Shuo Cai
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Catherine Zabkiewicz
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Chang Liu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| |
Collapse
|
5
|
Addante A, Roncero C, Lazcanoiturburu N, Méndez R, Almalé L, García-Álvaro M, ten Dijke P, Fabregat I, Herrera B, Sánchez A. A Signaling Crosstalk between BMP9 and HGF/c-Met Regulates Mouse Adult Liver Progenitor Cell Survival. Cells 2020; 9:cells9030752. [PMID: 32204446 PMCID: PMC7140668 DOI: 10.3390/cells9030752] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/17/2022] Open
Abstract
During chronic liver disease, hepatic progenitor cells (HPC, oval cells in rodents) become activated, proliferate, and differentiate into cholangiocytes and/or hepatocytes contributing to the final outcome of the regenerative process in a context-dependent fashion. Here, we analyze the crosstalk between the hepatocyte growth factor (HGF)/c-Met signaling axis, key for liver regeneration, and bone morphogenetic protein (BMP)9, a BMP family ligand that has emerged as a critical regulator of liver pathology. Our results show that HGF/c-Met signaling blocks BMP9-mediated apoptotic cell death, while it potentiates small mothers against decapentaplegic (SMAD)1 signaling triggered by BMP9 in oval cells. Interestingly, HGF-induced overactivation of SMAD1, -5, -8 requires the upregulation of TGF-β type receptor activin receptor-like kinase (ALK)1, and both ALK1 and SMAD1 are required for the counteracting effect of HGF on BMP9 apoptotic activity. On the other hand, we also prove that BMP9 triggers the activation of p38MAPK in oval cells, which drives BMP9-apoptotic cell death. Therefore, our data support a model in which BMP9 and HGF/c-Met signaling axes establish a signaling crosstalk via ALK1 that modulates the balance between the two pathways with opposing activities, SMAD1 (pro-survival) and p38 mitogen-activated protein kinases (p38MAPK; pro-apoptotic), which determines oval cell fate. These data help delineate the complex signaling network established during chronic liver injury and its impact on the oval cell regenerative response.
Collapse
Affiliation(s)
- Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Cesáreo Roncero
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Nerea Lazcanoiturburu
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Rebeca Méndez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Laura Almalé
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - María García-Álvaro
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), 08907 L’Hospitalet de Llobregat, Barcelona, Spain;
- School of Medicine and Health Sciences, University of Barcelona, 08007 Barcelona, Spain
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
- Correspondence: (B.H.); (A.S.); Tel.: +34 913941855 (A.S.)
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; (A.A.); (C.R.); (N.L.); (R.M.); (L.A.); (M.G.-Á.)
- Correspondence: (B.H.); (A.S.); Tel.: +34 913941855 (A.S.)
| |
Collapse
|
6
|
Jafri MA, Kalamegam G, Abbas M, Al-Kaff M, Ahmed F, Bakhashab S, Rasool M, Naseer MI, Sinnadurai V, Pushparaj PN. Deciphering the Association of Cytokines, Chemokines, and Growth Factors in Chondrogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells Using an ex vivo Osteochondral Culture System. Front Cell Dev Biol 2020; 7:380. [PMID: 32010693 PMCID: PMC6979484 DOI: 10.3389/fcell.2019.00380] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder associated with degradation and decreased production of the extracellular matrix, eventually leading to cartilage destruction. Limited chondrocyte turnover, structural damage, and prevailing inflammatory milieu prevent efficient cartilage repair and restoration of joint function. In the present study, we evaluated the role of secreted cytokines, chemokines, and growth factors present in the culture supernatant obtained from an ex vivo osteochondral model of cartilage differentiation using cartilage pellets (CP), bone marrow stem cells (BM-MSCs), and/or BM-MSCs + CP. Multiplex cytokine analysis showed differential secretion of growth factors (G-CSF, GM-CSF, HGF, EGF, VEGF); chemokines (MCP-1, MIP1α, MIP1β, RANTES, Eotaxin, IP-10), pro-inflammatory cytokines (IL-1β, IL-2, IL-5, IL-6, IL-8, TNFα, IL-12, IL-15, IL-17) and anti-inflammatory cytokines (IL-4, IL-10, and IL-13) in the experimental groups compared to the control. In silico analyses of the role of stem cells and CP in relation to the expression of various molecules, canonical pathways and hierarchical cluster patterns were deduced using the Ingenuity Pathway Analysis (IPA) software (Qiagen, United States). The interactions of the cytokines, chemokines, and growth factors that are involved in the cartilage differentiation showed that stem cells, when used together with CP, bring about a favorable cell signaling that supports cartilage differentiation and additionally helps to attenuate inflammatory cytokines and further downstream disease-associated pro-inflammatory pathways. Hence, the autologous or allogeneic stem cells and local cartilage tissues may be used for efficient cartilage differentiation and the management of OA.
Collapse
Affiliation(s)
- Mohammad Alam Jafri
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gauthaman Kalamegam
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis by Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia.,Faculty of Medicine, Asian Institute of Medicine, Science and Technology University, Bedong, Malaysia
| | - Mohammed Abbas
- Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis by Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Orthopaedic Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Al-Kaff
- Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis by Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Orthopaedic Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Farid Ahmed
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherin Bakhashab
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad Imran Naseer
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Vasan Sinnadurai
- Faculty of Medicine, Asian Institute of Medicine, Science and Technology University, Bedong, Malaysia
| | - Peter Natesan Pushparaj
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Wang X, Wang C, Gou W, Xu X, Wang Y, Wang A, Xu W, Guo Q, Liu S, Lu Q, Meng H, Yuan M, Peng J, Lu S. The optimal time to inject bone mesenchymal stem cells for fracture healing in a murine model. Stem Cell Res Ther 2018; 9:272. [PMID: 30359311 PMCID: PMC6202840 DOI: 10.1186/s13287-018-1034-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/27/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Bone marrow is an important source of stem cells, which can promote bone fracture healing. METHODS We investigated the optimal time to inject bone marrow mesenchymal stem cells (BMSCs) in a C57 murine unilateral, transverse, femur fracture model. BMSCs transfected with red fluorescent protein (RFP-BMSCs) were injected via the tail vein on day 1, 7, or 14 post-fracture. AMD3100 (inhibitor of stromal cell-derived factor 1 [SDF-1]) was also injected before RFP-BMSCs in one group for comparison; a control group received saline injections. RFP-BMSC migration and fracture healing were evaluated by in vivo fluorescence assay. Micro-CT was performed and mechanical testing and histological analysis. Chemokine levels were evaluated by quantitative real-time PCR and western blotting. RESULTS Following injection on day 7 post-fracture, RFP-BMSCs more frequently homed to the fracture site and remained for a longer duration. Bone volume and bone mineral density were increased when BMSCs were injected on day 7 post-fracture (P < 0.05). The mechanical properties of fractured femurs were improved following day-7 BMSC injection. Histology confirmed that BMSC injection improved the formation of new bones. CONCLUSIONS Chemokines that induce BMSC migration were highly expressed, and protein levels of osteogenesis-related factors were increased. Seven days after fracture may be the optimal time for injection of BMSCs to promote fracture healing. Additionally, the SDF-1/CXCR4 pathway may play an important role in fracture healing following BMSC injection.
Collapse
Affiliation(s)
- Xin Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Cheng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Wenlong Gou
- Department of Orthopaedics, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaolong Xu
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Aiyuan Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Wenjing Xu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Qiang Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Haoye Meng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Mei Yuan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Shibi Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Zhen R, Yang J, Wang Y, Li Y, Chen B, Song Y, Ma G, Yang B. Hepatocyte growth factor improves bone regeneration via the bone morphogenetic protein‑2‑mediated NF‑κB signaling pathway. Mol Med Rep 2018; 17:6045-6053. [PMID: 29436622 DOI: 10.3892/mmr.2018.8559] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/03/2017] [Indexed: 11/05/2022] Open
Abstract
Bone regeneration is an important process associated with the treatment of osteonecrosis, which is caused by various factors. Hepatocyte growth factor (HGF) is an active biological factor that has multifunctional roles in cell biology, life sciences and clinical medicine. It has previously been suggested that bone morphogenetic protein (BMP)‑2 exerts beneficial roles in bone formation, repair and angiogenesis in the femoral head. The present study aimed to investigate the benefits and molecular mechanisms of HGF in bone regeneration. The viability of osteoblasts and osteoclasts were studied in vitro. In addition, the expression levels of tumor necrosis factor (TNF)‑α, monocyte chemotactic protein (MCP)‑1, interleukin (IL)‑1 and IL‑6 were detected in a mouse fracture model following treatment with HGF. The expression and activity of nuclear factor (NF)‑κB were also analyzed in osteocytes post‑treatment with HGF. Histological analysis was used to determine the therapeutic effects of HGF on mice with fractures. The migration and differentiation of osteoblasts and osteoclasts were investigated in HGF‑incubated cells. Furthermore, angiogenesis and BMP‑2 expression were analyzed in the mouse fracture model post‑treatment with HGF. The results indicated that HGF regulates the cell viability of osteoblasts and osteoclasts, and also balanced the ratio between osteoblasts and osteoclasts. In addition, HGF decreased the serum expression levels of TNF‑α, MCP‑1, IL‑1 and IL‑6 in experimental mice. The results of a mechanistic analysis demonstrated that HGF upregulated p65, IκB kinase‑β and IκBα expression in osteoblasts from experimental mice. In addition, the expression levels of vascular endothelial growth factor, BMP‑2 receptor, receptor activator of NF‑κB ligand and macrophage colony‑stimulating factor were upregulated by HGF, which may effectively promote blood vessel regeneration, and contribute to the formation and revascularization of tissue‑engineered bone. Furthermore, HGF promoted BMP‑2 expression and enhanced angiogenesis at the fracture location. These results suggested that HGF treatment may significantly promote bone regeneration in a mouse fracture model. In conclusion, these results indicated that HGF is involved in bone regeneration, angiogenesis and the balance between osteoblasts and osteoclasts, thus suggesting that HGF may be considered a potential agent for the treatment of fractures via the promotion of bone regeneration through regulation of the BMP‑2‑mediated NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Ruixin Zhen
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| | - Jianing Yang
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| | - Yu Wang
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| | - Yubo Li
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| | - Bin Chen
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| | - Youxin Song
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| | - Guiyun Ma
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| | - Bo Yang
- Department of Spinal Surgery, Chengde Medical College Affiliated Hospital, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
9
|
Herrera B, Addante A, Sánchez A. BMP Signalling at the Crossroad of Liver Fibrosis and Regeneration. Int J Mol Sci 2017; 19:ijms19010039. [PMID: 29295498 PMCID: PMC5795989 DOI: 10.3390/ijms19010039] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Bone Morphogenetic Proteins (BMPs) belong to the Transforming Growth Factor-β (TGF-β) family. Initially identified due to their ability to induce bone formation, they are now known to have multiple functions in a variety of tissues, being critical not only during development for tissue morphogenesis and organogenesis but also during adult tissue homeostasis. This review focus on the liver as a target tissue for BMPs actions, devoting most efforts to summarize our knowledge on their recently recognized and/or emerging roles on regulation of the liver regenerative response to various insults, either acute or chronic and their effects on development and progression of liver fibrosis in different pathological conditions. In an attempt to provide the basis for guiding research efforts in this field both the more solid and more controversial areas of research were highlighted.
Collapse
Affiliation(s)
- Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| |
Collapse
|
10
|
Zabkiewicz C, Resaul J, Hargest R, Jiang WG, Ye L. Bone morphogenetic proteins, breast cancer, and bone metastases: striking the right balance. Endocr Relat Cancer 2017; 24:R349-R366. [PMID: 28733469 PMCID: PMC5574206 DOI: 10.1530/erc-17-0139] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/21/2017] [Indexed: 12/11/2022]
Abstract
Bone morphogenetic proteins (BMPs) belong to the TGF-β super family, and are essential for the regulation of foetal development, tissue differentiation and homeostasis and a multitude of cellular functions. Naturally, this has led to the exploration of aberrance in this highly regulated system as a key factor in tumourigenesis. Originally identified for their role in osteogenesis and bone turnover, attention has been turned to the potential role of BMPs in tumour metastases to, and progression within, the bone niche. This is particularly pertinent to breast cancer, which commonly metastasises to bone, and in which studies have revealed aberrations of both BMP expression and signalling, which correlate clinically with breast cancer progression. Ultimately a BMP profile could provide new prognostic disease markers. As the evidence suggests a role for BMPs in regulating breast tumour cellular function, in particular interactions with tumour stroma and the bone metastatic microenvironment, there may be novel therapeutic potential in targeting BMP signalling in breast cancer. This review provides an update on the current knowledge of BMP abnormalities and their implication in the development and progression of breast cancer, particularly in the disease-specific bone metastasis.
Collapse
Affiliation(s)
- Catherine Zabkiewicz
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Jeyna Resaul
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Rachel Hargest
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Wen Guo Jiang
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Lin Ye
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
11
|
Kim JW, Lee MN, Jeong BC, Oh SH, Kook MS, Koh JT. Chemical inhibitors of c-Met receptor tyrosine kinase stimulate osteoblast differentiation and bone regeneration. Eur J Pharmacol 2017; 806:10-17. [PMID: 28322831 DOI: 10.1016/j.ejphar.2017.03.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 01/19/2023]
Abstract
The c-Met receptor tyrosine kinase and its ligand, hepatocyte growth factor (HGF), have been recently introduced to negatively regulate bone morphogenetic protein (BMP)-induced osteogenesis. However, the effect of chemical inhibitors of c-Met receptor on osteoblast differentiation process has not been examined, especially the applicability of c-Met chemical inhibitors on in vivo bone regeneration. In this study, we demonstrated that chemical inhibitors of c-Met receptor tyrosine kinase, SYN1143 and SGX523, could potentiate the differentiation of precursor cells to osteoblasts and stimulate regeneration in calvarial bone defects of mice. Treatment with SYN1143 or SGX523 inhibited HGF-induced c-Met phosphorylation in MC3T3-E1 and C3H10T1/2 cells. Cell proliferation of MC3T3-E1 or C3H10T1/2 was not significantly affected by the concentrations of these inhibitors. Co-treatment with chemical inhibitor of c-Met and osteogenic inducing media enhanced osteoblast-specific genes expression and calcium nodule formation accompanied by increased Runx2 expression via c-Met receptor-dependent but Erk-Smad signaling independent pathway. Notably, the administration of these c-Met inhibitors significantly repaired critical-sized calvarial bone defects. Collectively, our results suggest that chemical inhibitors of c-Met receptor tyrosine kinase might be used as novel therapeutics to induce bone regeneration.
Collapse
Affiliation(s)
- Jung-Woo Kim
- Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Mi Nam Lee
- Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Byung-Chul Jeong
- Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Pharmacology, Seonam University Medical School, Namwon, Chonbuk 55724, Republic of Korea
| | - Sin-Hye Oh
- Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Min-Suk Kook
- Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Oral and Maxillofacial Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jeong-Tae Koh
- Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
12
|
Delitto D, Zhang D, Han S, Black BS, Knowlton AE, Vlada AC, Sarosi GA, Behrns KE, Thomas RM, Lu X, Liu C, George TJ, Hughes SJ, Wallet SM, Trevino JG. Nicotine Reduces Survival via Augmentation of Paracrine HGF-MET Signaling in the Pancreatic Cancer Microenvironment. Clin Cancer Res 2016; 22:1787-1799. [PMID: 26667487 PMCID: PMC4818664 DOI: 10.1158/1078-0432.ccr-15-1256] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 11/26/2015] [Indexed: 01/18/2023]
Abstract
PURPOSE The relationship between smoking and pancreatic cancer biology, particularly in the context of the heterogeneous microenvironment, remains incompletely defined. We hypothesized that nicotine exposure would lead to the augmentation of paracrine growth factor signaling between tumor-associated stroma (TAS) and pancreatic cancer cells, ultimately resulting in accelerated tumor growth and metastasis. EXPERIMENTAL DESIGN The effect of tobacco use on overall survival was analyzed using a prospectively maintained database of surgically resected patients with pancreatic cancer. Nicotine exposure was evaluated in vitro using primary patient-derived TAS and pancreatic cancer cells independently and in coculture. Nicotine administration was then assessed in vivo using a patient-derived pancreatic cancer xenograft model. RESULTS Continued smoking was associated with reduced overall survival after surgical resection. In culture, nicotine-stimulated hepatocyte growth factor (HGF) secretion in primary patient-derived TAS and nicotine stimulation was required for persistent pancreatic cancer cell c-Met activation in a coculture model. c-Met activation in this manner led to the induction of inhibitor of differentiation-1 (Id1) in pancreatic cancer cells, previously established as a mediator of growth, invasion and chemoresistance. HGF-induced Id1 expression was abrogated by both epigenetic and pharmacologic c-Met inhibition. In patient-derived pancreatic cancer xenografts, nicotine treatment augmented tumor growth and metastasis; tumor lysates from nicotine-treated mice demonstrated elevated HGF expression by qRT-PCR and phospho-Met levels by ELISA. Similarly, elevated levels of phospho-Met in surgically resected pancreatic cancer specimens correlated with reduced overall survival. CONCLUSIONS Taken together, these data demonstrate a novel, microenvironment-dependent paracrine signaling mechanism by which nicotine exposure promotes the growth and metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Dongyu Zhang
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Song Han
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Brian S Black
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Andrea E Knowlton
- Department of Periodontology and Oral Biology, University of Florida Health Science Center, Gainesville, Florida
| | - Adrian C Vlada
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - George A Sarosi
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida. North Florida/South Georgia Veterans Health System, University of Florida Health Science Center, Gainesville, Florida
| | - Kevin E Behrns
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Ryan M Thomas
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida. North Florida/South Georgia Veterans Health System, University of Florida Health Science Center, Gainesville, Florida
| | - Xiaomin Lu
- Department of Biostatistics and Children's Oncology Group, University of Florida Health Science Center, Gainesville, Florida
| | - Chen Liu
- Department of Pathology, Immunology, Laboratory Medicine, Colleges of Medicine, Dentistry and Public Health and Health Professions, University of Florida Health Science Center, Gainesville, Florida
| | - Thomas J George
- Department of Internal Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Steven J Hughes
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Shannon M Wallet
- Department of Periodontology and Oral Biology, University of Florida Health Science Center, Gainesville, Florida
| | - Jose G Trevino
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida.
| |
Collapse
|
13
|
Kim YH, Tabata Y. Dual-controlled release system of drugs for bone regeneration. Adv Drug Deliv Rev 2015; 94:28-40. [PMID: 26079284 DOI: 10.1016/j.addr.2015.06.003] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/23/2015] [Accepted: 06/08/2015] [Indexed: 02/08/2023]
Abstract
Controlled release systems have been noted to allow drugs to enhance their ability for bone regeneration. To this end, various biomaterials have been used as the release carriers of drugs, such as low-molecular-weight drugs, growth factors, and others. The drugs are released from the release carriers in a controlled fashion to maintain their actions for a long time period. Most research has been focused on the controlled release of single drugs to demonstrate the therapeutic feasibility. Controlled release of two combined drugs, so-called dual release systems, are promising and important for tissue regeneration. This is because the tissue regeneration process of bone formation is generally achieved by multiple bioactive molecules, which are produced from cells by other molecules. If two types of bioactive molecules, (i.e., drugs), are supplied in an appropriate fashion, the regeneration process of living bodies will be efficiently promoted. This review focuses on the bone regeneration induced by dual-controlled release of drugs. In this paper, various dual-controlled release systems of drugs aiming at bone regeneration are overviewed explaining the type of drugs and their release materials.
Collapse
|
14
|
Chen X, Song F, Jhamb D, Li J, Bottino MC, Palakal MJ, Stocum DL. The Axolotl Fibula as a Model for the Induction of Regeneration across Large Segment Defects in Long Bones of the Extremities. PLoS One 2015; 10:e0130819. [PMID: 26098852 PMCID: PMC4476796 DOI: 10.1371/journal.pone.0130819] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/26/2015] [Indexed: 12/25/2022] Open
Abstract
We tested the ability of the axolotl (Ambystoma mexicanum) fibula to regenerate across segment defects of different size in the absence of intervention or after implant of a unique 8-braid pig small intestine submucosa (SIS) scaffold, with or without incorporated growth factor combinations or tissue protein extract. Fractures and defects of 10% and 20% of the total limb length regenerated well without any intervention, but 40% and 50% defects failed to regenerate after either simple removal of bone or implanting SIS scaffold alone. By contrast, scaffold soaked in the growth factor combination BMP-4/HGF or in protein extract of intact limb tissue promoted partial or extensive induction of cartilage and bone across 50% segment defects in 30%-33% of cases. These results show that BMP-4/HGF and intact tissue protein extract can promote the events required to induce cartilage and bone formation across a segment defect larger than critical size and that the long bones of axolotl limbs are an inexpensive model to screen soluble factors and natural and synthetic scaffolds for their efficacy in stimulating this process.
Collapse
Affiliation(s)
- Xiaoping Chen
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Fengyu Song
- Department of Oral Biology, School of Dentistry, Indiana-University-Purdue University, Indianapolis, Indiana, United States of America
| | - Deepali Jhamb
- School of Informatics and Computing, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Jiliang Li
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Marco C. Bottino
- Department of Restorative Dentistry, Division of Dental Biomaterials, School of Dentistry, Indiana-University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Mathew J. Palakal
- School of Informatics and Computing, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - David L. Stocum
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
15
|
Abed E, Bouvard B, Martineau X, Jouzeau JY, Reboul P, Lajeunesse D. Elevated hepatocyte growth factor levels in osteoarthritis osteoblasts contribute to their altered response to bone morphogenetic protein-2 and reduced mineralization capacity. Bone 2015; 75:111-9. [PMID: 25667190 DOI: 10.1016/j.bone.2015.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/24/2015] [Accepted: 02/01/2015] [Indexed: 10/24/2022]
Abstract
PURPOSE Clinical and in vitro studies suggest that subchondral bone sclerosis due to abnormal osteoblasts is involved in the progression of osteoarthritis (OA). Human osteoblasts isolated from sclerotic subchondral OA bone tissue show an altered phenotype, a decreased canonical Wnt/ß-catenin pathway, and a reduced mineralization in vitro as well as in vivo. These alterations were linked with an abnormal response to BMP-2. OA osteoblasts release factors such as the hepatocyte growth factor (HGF) that contribute to cartilage loss whereas chondrocytes do not express HGF. HGF can stimulate BMP-2 expression in human osteoblasts, however, the role of HGF and its effect in OA osteoblasts remains unknown. Here we investigated whether elevated endogenous HGF levels in OA osteoblasts are responsible for their altered response to BMP-2. METHODS We prepared primary human subchondral osteoblasts using the sclerotic medial portion of the tibial plateaus of OA patients undergoing total knee arthroplasty, or from tibial plateaus of normal individuals obtained at autopsy. The expression of HGF was evaluated by qRT-PCR and the protein production by western blot analysis. HGF expression was reduced with siRNA technique whereas its activity was inhibited using the selective inhibitor PHA665752. Alkaline phosphatase activity (ALPase) and osteocalcin release were measured by substrate hydrolysis and EIA respectively. Canonical Wnt/β-catenin signaling (cWnt) was evaluated both by target gene expression using the TOPflash TCF/lef luciferase reporter assay and western blot analysis of β-catenin levels in response to Wnt3a stimulation. Mineralization in response to BMP-2 was evaluated by alizarin red staining. RESULTS The expression of HGF was increased in OA osteoblasts compared to normal osteoblasts and was maintained during their in vitro differentiation. OA osteoblasts released more HGF than normal osteoblasts as assessed by western blot analysis. HGF stimulated the expression of TGF-β1. BMP-2 dose-dependently (1 to 100 ng/ml) stimulated both ALPase and osteocalcin in normal osteoblasts whereas, it inhibited them in OA osteoblasts. HGF-siRNA treatments reversed this response in OA osteoblasts and restored the BMP-2 response. cWnt is reduced in OA osteoblasts compared to normal, and HGF-siRNA treatments increased cWnt in OA osteoblasts almost to normal. Smad1/5/8 phosphorylation in response to BMP-2, which is reduced in OA osteoblasts, was corrected when these cells were treated with PHA665752. The BMP-2-dependent mineralization of OA osteoblasts, which is also reduced compared to normal, was only partially restored by PHA665752 treatment whereas 28 days treatment with HGF reduced the mineralization of normal osteoblasts. CONCLUSION OA osteoblasts expressed more HGF than normal osteoblasts. Increased endogenous HGF production in OA osteoblasts stimulated the expression of TGF-β1 and reduced their response to BMP-2. Inhibiting HGF expression or HGF signaling restored the response to BMP-2 and Smad1/5/8 signaling. In addition, decreased HGF signaling partly corrects the abnormal mineralization of OA osteoblasts while increased HGF prevents the normal mineralization of normal osteoblasts. In summary, we hypothesize that sustained elevated HGF levels in OA osteoblasts drive their abnormal phenotype and is implicated in OA pathophysiology.
Collapse
Affiliation(s)
- E Abed
- Unité de recherche en Arthrose, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - B Bouvard
- UMR7365 IMoPA, Université de Lorraine/CNRS, Vandœuvre lès Nancy, 54505, France; Service de Rhumatologie, Centre Hospitalier Universitaire (CHU), Angers 49933, France
| | - X Martineau
- Unité de recherche en Arthrose, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - J-Y Jouzeau
- UMR7365 IMoPA, Université de Lorraine/CNRS, Vandœuvre lès Nancy, 54505, France; Service de Pharmacologie Clinique et de Toxicologie, Centre Hospitalier Universitaire (CHU), Nancy 54023, France
| | - P Reboul
- UMR7365 IMoPA, Université de Lorraine/CNRS, Vandœuvre lès Nancy, 54505, France
| | - D Lajeunesse
- Unité de recherche en Arthrose, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.
| |
Collapse
|
16
|
Yano K, Yasuda H, Takaoka K, Takahashi M, Nakamura H, Imai Y, Wakitani S. Fate, origin and roles of cells within free bone grafts. J Orthop Sci 2015; 20:390-6. [PMID: 25408483 DOI: 10.1007/s00776-014-0673-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 10/01/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND The efficacy of autologous bone grafting in repairing nonunion fractures, large bone defects and spinal instability is widely accepted. However, the cellular and molecular mechanisms underlying new bone formation in bone grafting have yet to be fully elucidated. The purpose of this study was to clarify the fate, origin and the contribution of the cells within the grafted bone. METHODS This study was designed to investigate the role and fate of cells contained in the grafted bone and their contribution to new bone formation in the graft in an animal model. Middiaphyseal cylindrical bone samples obtained from green fluorescent protein (GFP) transgenic and wild-type rats were transplanted into the back muscle of wild-type and GFP rats, respectively. The transplanted bones were evaluated by immunohistochemistry, in situ hybridization and quantitative reverse transcription polymerase chain reaction. RESULTS Immunohistochemical analyses showed that all the cells in the newly formed bone originated from the grafted bone, and osteoblasts were gradually replaced by host cells. Conversely, osteoclasts were immediately replaced by host cells 2 weeks after the bone graft. In addition, expression of bone morphogenetic protein (Bmp)-4, Bmp receptors and Noggin in the grafted bone was significantly upregulated before new bone formation occurred, indicating that the grafted cells might contribute to the recruitment of mesenchymal cells into the graft bed. CONCLUSION This study revealed the possible molecular mechanisms of the contribution of cells contained in grafted bone to facilitate new bone formation.
Collapse
Affiliation(s)
- Koichi Yano
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, Osaka, 545-8585, Japan
| | | | | | | | | | | | | |
Collapse
|
17
|
Masuda T, Otsu K, Kumakami-Sakano M, Fujiwara N, Ema M, Hitomi J, Sugiyama Y, Harada H. Combined Administration of BMP-2 and HGF Facilitate Bone Regeneration through Angiogenic Mechanisms. J HARD TISSUE BIOL 2015. [DOI: 10.2485/jhtb.24.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Tomoyuki Masuda
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University
- Division of Oral and Maxillofacial Surgery, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University
| | - Keishi Otsu
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University
| | - Mika Kumakami-Sakano
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University
| | - Naoki Fujiwara
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University
| | - Masatsugu Ema
- Research Center for Animal Life Science, Department of Stem Cells and Human Disease Models, Shiga University of Medical Science
| | - Jiro Hitomi
- Division of Human Embryology, Department of Anatomy, Iwate Medical University
| | - Yoshiki Sugiyama
- Division of Oral and Maxillofacial Surgery, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University
| | - Hidemitsu Harada
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University
| |
Collapse
|
18
|
Malhotra A, Pelletier M, Oliver R, Christou C, Walsh WR. Platelet-Rich Plasma and Bone Defect Healing. Tissue Eng Part A 2014; 20:2614-33. [DOI: 10.1089/ten.tea.2013.0737] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Angad Malhotra
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, The University of New South Wales, Prince of Wales Hospital, Randwick, Australia
| | - Matthew Pelletier
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, The University of New South Wales, Prince of Wales Hospital, Randwick, Australia
| | - Rema Oliver
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, The University of New South Wales, Prince of Wales Hospital, Randwick, Australia
| | - Chris Christou
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, The University of New South Wales, Prince of Wales Hospital, Randwick, Australia
| | - William R. Walsh
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, The University of New South Wales, Prince of Wales Hospital, Randwick, Australia
| |
Collapse
|
19
|
Durand M, Collombet JM, Frasca S, Begot L, Lataillade JJ, Le Bousse-Kerdilès MC, Holy X. In vivo hypobaric hypoxia performed during the remodeling process accelerates bone healing in mice. Stem Cells Transl Med 2014; 3:958-68. [PMID: 24944208 DOI: 10.5966/sctm.2013-0209] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We investigated the effects of respiratory hypobaric hypoxia on femoral bone-defect repair in mice because hypoxia is believed to influence both mesenchymal stromal cell (MSC) and hematopoietic stem cell mobilization, a process involved in the bone-healing mechanism. To mimic conditions of non-weight-bearing limb immobilization in patients suffering from bone trauma, our hypoxic mouse model was further subjected to hind-limb unloading. A hole was drilled in the right femur of adult male C57/BL6J mice. Four days after surgery, mice were subjected to hind-limb unloading for 1 week. Seven days after surgery, mice were either housed for 4 days in a hypobaric room (FiO2 at 10%) or kept under normoxic conditions. Unsuspended control mice were housed in either hypobaric or normoxic conditions. Animals were sacrificed on postsurgery day 11 to allow for collection of both contralateral and lesioned femurs, blood, and spleen. As assessed by microtomography, delayed hypoxia enhanced bone-healing efficiency by increasing the closing of the cortical defect and the newly synthesized bone volume in the cavity by +55% and +35%, respectively. Proteome analysis and histomorphometric data suggested that bone-repair improvement likely results from the acceleration of the natural bone-healing process rather than from extended mobilization of MSC-derived osteoprogenitors. Hind-limb unloading had hardly any effect beyond delayed hypoxia-enhanced bone-healing efficiency.
Collapse
Affiliation(s)
- Marjorie Durand
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Jean-Marc Collombet
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Sophie Frasca
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Laurent Begot
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Jean-Jacques Lataillade
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Marie-Caroline Le Bousse-Kerdilès
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| | - Xavier Holy
- Département Soutien Médico-Chirugical des Forces, Institut de Recherche Biomédicale des Armées (IRBA), Brétigny-sur-Orge, France; Centre de Transfusion Sanguine des Armées, Service de Recherche, Clamart, France; INSERM U972, Hôpital Paul Brousse, Villejuif, France
| |
Collapse
|
20
|
Malhotra A, Pelletier MH, Yu Y, Walsh WR. Can platelet-rich plasma (PRP) improve bone healing? A comparison between the theory and experimental outcomes. Arch Orthop Trauma Surg 2013. [PMID: 23197184 DOI: 10.1007/s00402-012-1641-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The increased concentration of platelets within platelet-rich plasma (PRP) provides a vehicle to deliver supra-physiologic concentrations of growth factors to an injury site, possibly accelerating or otherwise improving connective tissue regeneration. This potential benefit has led to the application of PRP in several applications; however, inconsistent results have limited widespread adoption in bone healing. This review provides a core understanding of the bone healing mechanisms, and corresponds this to the factors present in PRP. In addition, the current state of the art of PRP preparation, the key aspects that may influence its effectiveness, and treatment outcomes as they relate specifically to bone defect healing are presented. Although PRP does have a sound scientific basis, its use for bone healing appears only beneficial when used in combination with osteoconductive scaffolds; however, neither allograft nor autograft appear to be appropriate carriers. Aggressive processing techniques and very high concentrations of PRP may not improve healing outcomes. Moreover, many other variables exist in PRP preparation and use that influence its efficacy; the effect of these variables should be understood when considering PRP use. This review includes the essentials of what has been established, what is currently missing in the literature, and recommendations for future directions.
Collapse
Affiliation(s)
- Angad Malhotra
- Surgical and Orthopaedic Research Laboratories, Prince of Wales Clinical School, The University of New South Wales, Sydney, Australia.
| | | | | | | |
Collapse
|
21
|
Goshima K, Nakase J, Xu Q, Matsumoto K, Tsuchiya H. Repair of segmental bone defects in rabbit tibia promoted by a complex of β-tricalcium phosphate and hepatocyte growth factor. J Orthop Sci 2012; 17:639-48. [PMID: 22763716 DOI: 10.1007/s00776-012-0262-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 06/01/2012] [Indexed: 01/03/2023]
Abstract
BACKGROUND Segmental bone defect repair remains a clinical and scientific challenge with increasing interest focused on bone tissue engineering. Clinical studies are ongoing to address application of hepatocyte growth factor (HGF) for treatment of some diseases; however, the use of HGF in bone tissue engineering has not been addressed. This study was performed to evaluate the effect of HGF in a complex of β-tricalcium phosphate (β-TCP) and collagen in repairing segmental bone defects. METHODS Segmental bone defects 5 mm long were created in the middle of the tibial shafts of rabbits. The defect was stabilized with external fixators and implanted with a complex of β-TCP granules and collagen, with or without 100 μg recombinant human HGF. Biweekly, bone regeneration and β-TCP resorption were assessed radiographically and histologically. At 4 and 8 weeks, bone regeneration was evaluated by use of micro-computed tomography and mechanical tests. RESULTS Compared with the bone tissue treated with β-TCP and collagen, mineralization, angiogenesis, new bone formation, and absorption of β-TCP were promoted 4 weeks postoperatively by treatment with HGF in the β-TCP and collagen group. These changes were associated with promoting biomechanical regeneration. By 8 weeks, the formation of bone marrow in newly generated bone and absorption of the β-TCP granules were completed in a shorter period by combining HGF with β-TCP and collagen, compared with tissues without HGF. CONCLUSIONS The combined application of HGF in a β-TCP and collagen matrix promoted histological bone healing and augmented mechanical strength of the healing bone, particularly in the early stages. The combined use of HGF and β-TCP for treatment of bone defects made a substantial difference.
Collapse
Affiliation(s)
- Kenichi Goshima
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan.
| | | | | | | | | |
Collapse
|
22
|
Yoshida S, Iwasaki R, Kawana H, Miyauchi Y, Hoshi H, Miyamoto H, Mori T, Kanagawa H, Katsuyama E, Fujie A, Hao W, Kobayashi T, Sato Y, Miyamoto K, Morioka H, Matsumoto M, Chiba K, Toyama Y, Nakagawa T, Miyamoto T. PDGFBB promotes PDGFRα-positive cell migration into artificial bone in vivo. Biochem Biophys Res Commun 2012; 421:785-9. [DOI: 10.1016/j.bbrc.2012.04.084] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 04/14/2012] [Indexed: 12/01/2022]
|
23
|
Nakase J, Kitaoka K, Matsumoto K, Tomita K. Facilitated tendon-bone healing by local delivery of recombinant hepatocyte growth factor in rabbits. Arthroscopy 2010; 26:84-90. [PMID: 20117631 DOI: 10.1016/j.arthro.2009.06.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 04/12/2009] [Accepted: 06/27/2009] [Indexed: 02/02/2023]
Abstract
PURPOSE This study was performed to evaluate the therapeutic effect of hepatocyte growth factor (HGF) on tendon-bone healing in a rabbit model. METHODS In adult rabbits the long digital extensor tendon was detached from the lateral femoral condyle, and the free end of the tendon was inserted into a tunnel drilled into the proximal tibial metaphysis. Cancellous bone obtained during drilling of the tibial hole was soaked in saline solution or solution containing 100-microg/mL human recombinant HGF and then transplanted into the bone tunnel. Junctional healing between the tendon and the bone was evaluated by histologic analysis and uniaxial load-to-failure testing at 2, 4, 6, 8, and 12 weeks after surgery. RESULTS In the saline solution-treated control group, Sharpey-like fibers, which connected the tendon graft and the bone tissue, appeared 6 weeks after treatment. At 8 weeks after treatment, maturation of lamellar bone was seen, and at 12 weeks, the adhesion between tendon and bone appeared to be supported by indirect insertion of fibrocartilaginous tissue, wherein the border between the fibrocartilaginous tissue and tendon or bone was significant. In the HGF-treated group, the fibrous tissues were parallel to the load axis, and lamellar bone and Sharpey-like fibers appeared as early as 4 weeks after treatment. At 12 weeks, junctional tissue, characterized by a continuous 4-layer structure of bone, calcified cartilage, fibrocartilage, and tendon, was regenerated by a direct insertion. On biomechanical testing, the HGF-treated group had significantly better biomechanical properties than the control group at 2 and 4 weeks. The histologic improvement caused by HGF treatment was associated with the biomechanical improvement. CONCLUSIONS Local administration of recombinant HGF promotes the adhesive healing process at the tendon-bone junction, both histologically and mechanically, after ligament reconstruction in a rabbit model. CLINICAL RELEVANCE Application of HGF may be considered as a new therapeutic approach to accelerate healing and rehabilitation after ligament reconstruction.
Collapse
Affiliation(s)
- Junsuke Nakase
- Department of Orthopaedic Surgery, School of Medicine, Kanazawa University, Kanazawa, Japan.
| | | | | | | |
Collapse
|
24
|
Kawasaki T, Niki Y, Miyamoto T, Horiuchi K, Matsumoto M, Aizawa M, Toyama Y. The effect of timing in the administration of hepatocyte growth factor to modulate BMP-2-induced osteoblast differentiation. Biomaterials 2009; 31:1191-8. [PMID: 19913294 DOI: 10.1016/j.biomaterials.2009.10.048] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 10/20/2009] [Indexed: 12/21/2022]
Abstract
Development of bone morphogenetic protein (BMP) signaling modulators may provide useful therapeutic options for the treatment of large bony defects in clinical settings. Controversy remains over whether hepatocyte growth factor (HGF) is a positive or negative modulator of BMP-induced osteogenesis. This study analyzed osteogenic properties of HGF, particularly during BMP-2-induced bone formation. Using a mouse model of ectopic bone formation, HGF-impregnated gelatin sponges displayed significantly reduced bone formation induced by BMP-2, both radiologically and histologically. Abrogation of endogenous HGF production by knockdown of HGF mRNA resulted in upregulation of BMP-2-induced ALP activity for C2C12 myoblasts in vitro. In contrast, addition of exogenous HGF inhibited BMP-2-induced ALP activity and osteocalcin production by mouse embryonic fibroblasts (MEFs) through HGF-c-Met interactions. Inhibition of ALP activity by HGF was rescued by U0126, a MEK1/2 inhibitor, indicating that HGF suppresses the BMP-2-Smad axis via activation of ERK1/2. Importantly, treatment with HGF prior to administration of BMP-2 induced cellular proliferation of MEFs and did not influence subsequent osteoblast differentiation induced by BMP-2. The effects of HGF may differ according to the differentiation stage of mesenchymal stem cells, which would explain the inconsistencies seen in osteogenic properties of HGF in previous reports. The timing of HGF treatment is critical and should be carefully determined for successful induction of bone formation by BMPs.
Collapse
Affiliation(s)
- Toshiki Kawasaki
- Department of Orthopaedic Surgery, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
25
|
Shen Q, Little SC, Xu M, Haupt J, Ast C, Katagiri T, Mundlos S, Seemann P, Kaplan FS, Mullins MC, Shore EM. The fibrodysplasia ossificans progressiva R206H ACVR1 mutation activates BMP-independent chondrogenesis and zebrafish embryo ventralization. J Clin Invest 2009; 119:3462-72. [PMID: 19855136 DOI: 10.1172/jci37412] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 08/25/2009] [Indexed: 12/30/2022] Open
Abstract
Patients with classic fibrodysplasia ossificans progressiva, a disorder characterized by extensive extraskeletal endochondral bone formation, share a recurrent mutation (R206H) within the glycine/serine-rich domain of ACVR1/ALK2, a bone morphogenetic protein type I receptor. Through a series of in vitro assays using several mammalian cell lines and chick limb bud micromass cultures, we determined that mutant R206H ACVR1 activated BMP signaling in the absence of BMP ligand and mediated BMP-independent chondrogenesis that was enhanced by BMP. We further investigated the interaction of mutant R206H ACVR1 with FKBP1A, a glycine/serine domain-binding protein that prevents leaky BMP type I receptor activation in the absence of ligand. The mutant protein exhibited reduced binding to FKBP1A in COS-7 simian kidney cell line assays, suggesting that increased BMP pathway activity in COS-7 cells with R206H ACVR1 is due, at least in part, to decreased binding of this inhibitory factor. Consistent with these findings, in vivo analyses of zebrafish embryos showed BMP-independent hyperactivation of BMP signaling in response to the R206H mutant, resulting in increased embryonic ventralization. These data support the conclusion that the mutant R206H ACVR1 receptor in FOP patients is an activating mutation that induces BMP signaling in a BMP-independent and BMP-responsive manner to promote chondrogenesis, consistent with the ectopic endochondral bone formation in these patients.
Collapse
Affiliation(s)
- Qi Shen
- Department of Orthopaedic Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6081, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Nakao Y, Koike T, Ohta Y, Manaka T, Imai Y, Takaoka K. Parathyroid hormone enhances bone morphogenetic protein activity by increasing intracellular 3', 5'-cyclic adenosine monophosphate accumulation in osteoblastic MC3T3-E1 cells. Bone 2009; 44:872-7. [PMID: 19442611 DOI: 10.1016/j.bone.2009.01.370] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2008] [Revised: 01/14/2009] [Accepted: 01/21/2009] [Indexed: 12/11/2022]
Abstract
Intermittent subcutaneous injections of parathyroid hormone (PTH) increase bone mass in a variety of animal models and humans. The anabolic actions of PTH on osteogenic cells are mainly mediated through the protein kinase A (PKA) signaling pathway via PTH receptor 1 (PTHR1). We have already reported 3', 5'-cyclic adenosine monophosphate (cAMP)/PKA-mediated enhancement of bone morphogenetic protein (BMP) signaling. Herein, we focused on the involvement of PTH in BMP signaling pathways in the MC3T3-E1 mouse osteoblastic cell line, to elucidate a potential mechanism of the anabolic actions of PTH on bone formation. Elevation of intracellular cAMP level in MC3T3-E1 cells by addition of PTH (10(-7) M) to culture media was transient without significant effect on biological actions of BMP. Cyclic addition of PTH (10 cyclic additions of 10(-8) M PTH at 3-min intervals) maintained a high intracellular cAMP level for about 2 h and mRNA expression and enzymatic activity of alkaline phosphatase (ALP) by BMP was enhanced by this addition. Relative luciferase expression assay in MC3T3-E1 cells using the Id1 promoter, an early response gene to BMPs, enhanced elevation of transcriptional activity in response to recombinant human BMP-2 by concomitant addition of PTH and BMP. Furthermore, cyclic PTH treatment significantly further suppressed BMP-induced inhibitory Smad6 expression. H89 (PKA inhibitor) almost completely abolished PTH actions on BMP signaling. IBMX (phosphodiesterase inhibitor) enhanced PTH actions. These results suggest that PTH enhances BMP signaling when PTH-induced intracellular cAMP level is maintained for a few hours, accelerating BMP actions to promote osteoblastic function and anabolic actions of new bone formation.
Collapse
Affiliation(s)
- Yoshihiro Nakao
- Department of Orthopaedic Surgery, Osaka City University Medical School, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Takayama K, Suzuki A, Manaka T, Taguchi S, Hashimoto Y, Imai Y, Wakitani S, Takaoka K. RNA interference for noggin enhances the biological activity of bone morphogenetic proteins in vivo and in vitro. J Bone Miner Metab 2009; 27:402-11. [PMID: 19252814 DOI: 10.1007/s00774-009-0054-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 10/07/2008] [Indexed: 11/29/2022]
Abstract
Noggin is a major extracellular antagonist to bone morphogenetic proteins (BMPs) which binds to BMPs and blocks binding of them to BMP-specific receptors and negatively regulates BMP-induced osteoblastic differentiation. In this study, we investigated the effect of noggin silencing by transfection of small interfering RNA (siRNA) on BMP-induced osteoblastic differentiation in vitro and ectopic bone formation in vivo induced by recombinant human BMP-2 (rhBMP-2). Noggin mRNA expression was up-regulated in response to rhBMP-2 in C2C12 cells, a myoblastic cell line, in dose- and time-dependent fashion as determined by real-time RT-PCR assay. Silencing of noggin expression by transfection of noggin siRNA suppressed BMP-stimulated noggin expression, resulting in acceleration of BMP-induced osteoblastic differentiation. For in vivo noggin silencing, siRNA was injected locally into back muscles and transfected into local cells by electroporation, where rhBMP-2-retaining (5 microg) collagen disks had been surgically placed. The implants were harvested at 2 weeks after surgery from experimental and control group mice and analyzed by radiological and histological methods. As a result, bone mineral content of ossicles ectopically induced by rhBMP-2 was significantly increased by silencing of noggin. Our findings suggest that silencing of noggin enhances the osteoblastic differentiation of BMP-responding cells in vitro and new bone formation induced by rhBMP-2 in vivo by eliminating negative regulation of the effects of BMP. RNA interference might be useful for intensifying the effects of BMP in promoting new bone (callus) formation in repair of damaged bone.
Collapse
Affiliation(s)
- Kazushi Takayama
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abenoku, Osaka, 545-8585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Matsubara H, Tsuchiya H, Watanabe K, Takeuchi A, Tomita K. Percutaneous nonviral delivery of hepatocyte growth factor in an osteotomy gap promotes bone repair in rabbits: a preliminary study. Clin Orthop Relat Res 2008; 466:2962-72. [PMID: 18813894 PMCID: PMC2628245 DOI: 10.1007/s11999-008-0493-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2008] [Accepted: 08/19/2008] [Indexed: 01/31/2023]
Abstract
Hepatocyte growth factor (HGF) was initially identified in cultured hepatocytes and subsequently reported to induce angiogenic, morphogenic, and antiapoptotic activity in various tissues. These properties suggest a potential influence of HGF on bone healing. We asked if gene transfer of human HGF (hHGF) into an osteotomy gap with a hemagglutinating virus of Japan-envelope (HVJ-E) vector promotes bone healing in rabbits. HVJ-E that contained either hHGF or control plasmid was percutaneously injected into the osteotomy gap of rabbit tibias on Day 14. The osteotomy gap was evaluated by radiography, pQCT, mechanical tests, and histology at Week 8. The expression of hHGF was evaluated by reverse transcriptase-polymerase chain reaction and immunohistochemistry at Week 3. Radiography, pQCT, and histology suggested the hHGF group had faster fracture healing. Mechanical tests demonstrated the hHGF group had greater mechanical strength. The injected tissues at 3 weeks expressed hHGF mRNA by reverse transcriptase-polymerase chain reaction. hHGF-positive immunohistochemical staining was observed in various cells at the osteotomy gap at Week 3. The data suggest delivery of hHGF plasmid into the osteotomy gap promotes fracture repair, and HGF could become a novel agent for fracture treatment.
Collapse
Affiliation(s)
- Hidenori Matsubara
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Koji Watanabe
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Akihiko Takeuchi
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Katsuro Tomita
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| |
Collapse
|
29
|
Su N, Yang J, Xie Y, Du X, Lu X, Yin Z, Yin L, Qi H, Zhao L, Feng J, Chen L. Gain-of-function mutation of FGFR3 results in impaired fracture healing due to inhibition of chondrocyte differentiation. Biochem Biophys Res Commun 2008; 376:454-9. [PMID: 18789890 DOI: 10.1016/j.bbrc.2008.08.165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2008] [Accepted: 08/28/2008] [Indexed: 11/15/2022]
Abstract
Fracture healing is a complicated regeneration process which to some extent recapitulates bone development. Fibroblast growth factor receptor 3 (FGFR3) has a negative regulatory effect on endochondral ossification, and FGFR3 is also expressed in prehypertrophic and hypertrophic chondrocytes during fracture healing. However, the actual role of FGFR3 during bone regeneration is not fully understood. Therefore we investigated the role of FGFR3 in fracture repair using a non-stabilized fracture model. Fracture repair in gain-of-function mutation of FGFR3 (Fgfr3(G369C/+)) mice was delayed, with more cartilage callus on day 14 and residue of cartilage in the callus on day 21. Histologic, in-situ hybridization and qRT-PCR analysis showed that differentiation of mesenchymal cells into chondrocytes and hypertrophic differentiation was delayed in Fgfr3(G369C/+) mice during fracture healing. These results indicated that activating mutation of FGFR3 could lead to impaired bone repair due to inhibition of chondrocyte differentiation.
Collapse
Affiliation(s)
- Nan Su
- State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Khan SN, Solaris J, Ramsey KE, Yang X, Bostrom MP, Stephan D, Daluiski A. Identification of novel gene expression in healing fracture callus tissue by DNA microarray. HSS J 2008; 4:149-60. [PMID: 18752025 PMCID: PMC2553169 DOI: 10.1007/s11420-008-9087-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Accepted: 06/23/2008] [Indexed: 02/07/2023]
Abstract
Fracture healing requires controlled expression of thousands of genes. Only a small fraction of these genes have been isolated and fewer yet have been shown to play a direct role in fracture healing. The purpose of this study was threefold: (1) to develop a reproducible open femur model of fracture healing that produces consistent fracture calluses for subsequent RNA extraction, (2) to use this model to determine temporal expression patterns of known and unknown genes using DNA microarray expression profiling, and (3) to identify and validate novel gene expression in fracture healing. In the initial arm of the study, a total of 56 wild-type C57BL/6 mice were used. An open, stabilized diaphyseal femur fracture was created. Animals were killed at 1, 5, 7, 10, 14, 21, and 35 days after surgery and the femurs were harvested for analysis. At each time point, fractures were radiographed and sectioned for histologic analyses. Tissue from fracture callus at all stages following fracture yielded reproducibly large amounts of mRNA. Expression profiling revealed that genes cluster by function in a manner similar to the histologic stages of fracture healing. Based on the expression profiling of fracture tissue, temporal expression patterns of several genes known to be involved in fracture healing were verified. Novel expression of multiple genes in fracture callous tissue was also revealed including leptin and leptin receptor. In order to test whether leptin signaling is required for fracture repair, mice deficient in leptin or its receptor were fractured using the same model. Fracture calluses of mice deficient in both leptin or leptin receptor are larger than wild-type mice fractures, likely due to a delay in mineralization, revealing a previously unrecognized role of leptin signaling in fracture healing. This novel model of murine fracture repair is useful in examining both global changes in gene expression as well as individual signaling pathways, which can be used to identify specific molecular mechanisms of fracture healing.
Collapse
Affiliation(s)
- Safdar N. Khan
- Department of Orthopaedic Surgery, University of California, 4860 Y Street, Suite 1700, Davis, Sacramento, CA 95817 USA
| | - Jorge Solaris
- The Hospital for Special Surgery, 523 E 72nd Street, New York, NY 10021 USA
| | - Keri E. Ramsey
- Translational Genomics Research Institute, 445 N. Fifth Street, Phoenix, AZ 85004 USA
| | - Xu Yang
- The Hospital for Special Surgery, 523 E 72nd Street, New York, NY 10021 USA
| | | | - Dietrich Stephan
- Translational Genomics Research Institute, 445 N. Fifth Street, Phoenix, AZ 85004 USA
| | - Aaron Daluiski
- The Hospital for Special Surgery, 523 E 72nd Street, New York, NY 10021 USA
| |
Collapse
|
31
|
Silva GA, Coutinho OP, Ducheyne P, Reis RL. Materials in particulate form for tissue engineering. 2. Applications in bone. J Tissue Eng Regen Med 2008; 1:97-109. [PMID: 18038398 DOI: 10.1002/term.1] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Materials in particulate form have been the subjects of intensive research in view of their use as drug delivery systems. While within this application there are still issues to be addressed, these systems are now being regarded as having a great potential for tissue engineering applications. Bone repair is a very demanding task, due to the specific characteristics of skeletal tissues, and the design of scaffolds for bone tissue engineering presents several difficulties. Materials in particulate form are now seen as a means of achieving higher control over parameters such as porosity, pore size, surface area and the mechanical properties of the scaffold. These materials also have the potential to incorporate biologically active molecules for release and to serve as carriers for cells. It is believed that the combination of these features would create a more efficient approach towards regeneration. This review focuses on the application of materials in particulate form for bone tissue engineering. A brief overview of bone biology and the healing process is also provided in order to place the application in its broader context. An original compilation of molecules with a documented role in bone tissue biology is listed, as they have the potential to be used in bone tissue engineering strategies. To sum up this review, examples of works addressing the above aspects are presented.
Collapse
Affiliation(s)
- G A Silva
- 3Bs Research Group--Biomaterials, Biodegradables, Biomimetics-University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | | | | | | |
Collapse
|
32
|
Nakagawa K, Imai Y, Ohta Y, Takaoka K. Prostaglandin E2 EP4 agonist (ONO-4819) accelerates BMP-induced osteoblastic differentiation. Bone 2007; 41:543-8. [PMID: 17681894 DOI: 10.1016/j.bone.2007.06.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 06/03/2007] [Accepted: 06/20/2007] [Indexed: 10/23/2022]
Abstract
Bone morphogenetic proteins (BMPs) were originally isolated based on their ability to induce ectopic cartilage and bone formation. The agents to promote the local bone formation with BMP would be beneficial to promote bone repair and to shorten the treatment period. For this purpose, we have examined ONO-4819, which is a prostaglandin (PG) E2 EP4 receptor selective agonist (EP4A), as a positive modulators for the efficacy of BMPs. In our previous study, the systemic and local (with biodegradable synthetic polymers) administration of EP4A led to a significant augmentation of ossicle mass. But the mechanisms how EP4A accelerates the BMP-mediated bone formation are still unknown. In this study, we have examined how EP4A facilitates the BMP signaling using in vitro system with pluripotent stromal cell line, ST2. The mRNA expressions of Osterix and ALP (a marker enzyme of osteoblastic differentiation) and enzymatic activity of ALP in the ST2 cells were elevated significantly by BMP treatment. This elevation was further elevated by addition of the EP4A. The accelerated BMP action by the EP4A was abolished by pre-treatment with PKA inhibitor. This study suggests that ONO-4819 accelerates BMP-induced osteoblastic differentiation of ST2 cells by stimulating the commitment for osteoblastic lineage. Thus PKA signaling pathway would be the main intracellular signaling pathway of the EP4 for the anabolic effect of bone and mineral metabolisms.
Collapse
Affiliation(s)
- Keisuke Nakagawa
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Asahimachi 1-4-3, Osaka 545-8585, Japan.
| | | | | | | |
Collapse
|
33
|
Gruber R, Koch H, Doll BA, Tegtmeier F, Einhorn TA, Hollinger JO. Fracture healing in the elderly patient. Exp Gerontol 2006; 41:1080-93. [PMID: 17092679 DOI: 10.1016/j.exger.2006.09.008] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 09/11/2006] [Accepted: 09/12/2006] [Indexed: 01/13/2023]
Abstract
Clinical experience gives rise to the impression that there are differences in fracture healing in different age groups. It is evident that fractures heal more efficiently in children than in adults. However, minimal objective knowledge exists to evaluate this assumption. Temporal, spatial, and cellular quantitative and qualitative interrelationships, as well as signaling molecules and extracellular matrix have not been comprehensively and adequately elucidated for fracture healing in the geriatric skeleton. The biological basis of fracture healing will provide a context for revealing the pathophysiology of delayed or even impaired bone regeneration in the elderly. We will summarize experimental studies on age-related changes at the cellular and molecular level that will add to the pathophysiological understanding of the compromised bone regeneration capacity believed to exist in the elderly patient. We will suggest why this understanding would be useful for therapeutics focused on bone regeneration, in particular fracture healing at an advanced age.
Collapse
Affiliation(s)
- Reinhard Gruber
- Department of Oral Surgery, Medical University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
34
|
Kawase T, Okuda K, Yoshie H. A Hepatocyte Growth Factor (HGF)/receptor autocrine loop regulates constitutive self-renewal of human periodontal ligament cells but reduces sensitivity to exogenous HGF. J Periodontol 2006; 77:1723-30. [PMID: 17032116 DOI: 10.1902/jop.2006.060031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND In addition to its prominent role in liver regeneration, hepatocyte growth factor (HGF) is now generally thought to be produced by mesenchymal cells to promote the regeneration of epithelial tissue by a paracrine mechanism. However, it is not known how or if HGF could be involved in the regeneration of periodontal tissues. The purpose of this study was to characterize the ability of normal human periodontal ligament (PDL) cells to produce or respond to HGF. METHODS PDL cells derived from healthy young volunteers were used from passages four through 10. HGF receptors were detected both by immunocytochemical staining and Western-blotting analysis. Both DNA synthesis (by bromo-deoxyuridine [BrdU]-incorporation) and secreted HGF were quantified by enzyme-linked immunosorbent assays. Mitogen-activated protein kinase (MAPK) phosphorylation was also analyzed by Western blot. RESULTS Despite the immunocytochemical demonstration of HGF receptor protein in the cytoplasm and on the plasma membrane of PDL cells, exogenous recombinant human HGF did not exert the mitogenic effects expected. As reported for other mesenchymal cells, PDL cells were found to secrete HGF. Treatments with neutralizing anti-HGF antibody significantly suppressed constitutive PDL cell proliferation and sustained the receptor protein at higher levels than in non-treated cells. Under these conditions, exogenous HGF rapidly phosphorylated extracellular signal-regulated kinase (ERK), an action linked to the cell proliferation and downregulation of cell-surface receptors. CONCLUSIONS Unlike other known mesenchymal or epithelial cells, these findings suggest that normal PDL cells from young donors possess a constitutive HGF/receptor autocrine loop that normally regulates their replacement self-proliferation but reduces sensitivity to exogenously applied HGF by acute receptor downregulation.
Collapse
Affiliation(s)
- Tomoyuki Kawase
- Division of Oral Regeneration and Transplantation Research, Department of Tissue Regeneration and Reconstruction, Institute of Medicine and Dentistry, Niigata University, Niigata, Japan.
| | | | | |
Collapse
|
35
|
Nakase T, Yoshikawa H. Potential roles of bone morphogenetic proteins (BMPs) in skeletal repair and regeneration. J Bone Miner Metab 2006; 24:425-33. [PMID: 17072733 DOI: 10.1007/s00774-006-0718-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Accepted: 07/20/2006] [Indexed: 12/20/2022]
Affiliation(s)
- Takanobu Nakase
- Department of Orthopaedic Surgery, Hoshigaokakoseinenkin Hospital, 4-8-1 Hoshioka, Hirakata, Osaka, 573-8511, Japan.
| | | |
Collapse
|