51
|
Abnormal inhibition of osteoclastogenesis by mesenchymal stem cells through the miR-4284/CXCL5 axis in ankylosing spondylitis. Cell Death Dis 2019; 10:188. [PMID: 30804325 PMCID: PMC6389901 DOI: 10.1038/s41419-019-1448-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/24/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022]
Abstract
Ankylosing spondylitis (AS) is a common inflammatory autoimmune disease, characterized by pathological osteogenesis. Mesenchymal stem cells (MSCs), as the main source of osteoblasts, participate in bone remodeling not only through differentiation into osteoblasts but also through indirect regulation of osteoclastogenesis. Our previous study indicated that the stronger osteogenic differentiation of MSCs from AS patients (ASMSCs) involved in pathological osteogenesis. However, whether there is any abnormality in the regulation of osteoclastogenesis by ASMSCs remains unclear. In this study, ASMSCs or MSCs from healthy donors (HDMSCs) were co-cultured with CD14 + monocytes in osteoclast induction medium. Our results demonstrated that ASMSCs exhibited a stronger capacity to inhibit osteoclastogenesis than HDMSCs. To explore underlying mechanisms, cytokine array assays were performed, showing that ASMSCs secreted more CXCL5 than HDMSCs, which was confirmed by enzyme-linked immunosorbent assays. Moreover, inhibition of osteoclastogenesis by ASMSCs was recovered by decreasing CXCL5. Besides, the inhibitory effect of CXCL5 on osteoclastogenesis was confirmed by exogenous addition. Bioinformatics analysis was applied to find the interaction between miR-4284 and CXCL5, which was verified by luciferase reporter assays. Furthermore, we used miR-4284 inhibitors or mimics to prove that the expression of CXCL5 was regulated by miR-4284. Further analysis showed that downregulation of miR-4284 in MSCs resulted in increase of CXCL5, markedly inhibiting osteoclastogenesis, whereas upregulation of miR-4284 in MSCs had the opposite effect. Our findings indicate that ASMSCs exhibit a stronger capacity to inhibit osteoclastogenesis than HDMSCs through the miR-4284/CXCL5 axis, which provide a new perspective on the mechanism of pathologic osteogenesis in AS.
Collapse
|
52
|
Abstract
A history of prior fracture is the most reliable indicator of prospective fracture risk. Increased fracture risk is not confined to the region of the prior fracture, but is operant at all skeletal sites, providing strong evidence of systemic bone loss after fracture. Animal and human studies suggest that systemic bone loss begins shortly after fracture and persists for several years in humans. In fact, bone quantity and bone quality may never fully return to their pre-fracture levels, especially in older subjects, demonstrating a need for improved understanding of the mechanisms leading to systemic bone loss after fracture in order to reduce subsequent fracture risk. Although the process remains incompletely understood, mechanical unloading (disuse), systemic inflammation, and hormones that control calcium homeostasis may all contribute to systemic bone loss. Additionally, individual factors can potentially affect the magnitude and time course of systemic bone loss and recovery. The magnitude of systemic bone loss correlates positively with injury severity and age. Men may also experience greater bone loss or less recovery than women after fracture. This review details the current understanding of systemic bone loss following fracture, including possible underlying mechanisms and individual factors that may affect this injury response.
Collapse
|
53
|
Hsu WB, Hsu WH, Hung JS, Shen WJ, Hsu RWW. Transcriptome analysis of osteoblasts in an ovariectomized mouse model in response to physical exercise. Bone Joint Res 2018; 7:601-608. [PMID: 30581558 PMCID: PMC6269594 DOI: 10.1302/2046-3758.711.bjr-2018-0075.r2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Objectives Osteoporosis is a metabolic disease resulting in progressive loss of bone mass as measured by bone mineral density (BMD). Physical exercise has a positive effect on increasing or maintaining BMD in postmenopausal women. The contribution of exercise to the regulation of osteogenesis in osteoblasts remains unclear. We therefore investigated the effect of exercise on osteoblasts in ovariectomized mice. Methods We compared the activity of differentially expressed genes of osteoblasts in ovariectomized mice that undertook exercise (OVX+T) with those that did not (OVX), using microarray and bioinformatics. Results Many inflammatory pathways were significantly downregulated in the osteoblasts after exercise. Meanwhile, IBSP and SLc13A5 gene expressions were upregulated in the OVX+T group. Furthermore, in in vitro assay, IBSP and SLc13A5 mRNAs were also upregulated during the osteogenic differentiation of MC3T3-E1 and 7F2 cells. Conclusion These findings suggest that exercise may not only reduce the inflammatory environment in ovariectomized mice, indirectly suppressing the overactivated osteoclasts, but may also directly activate osteogenesis-related genes in osteoblasts. Exercise may thus prevent the bone loss caused by oestrogen deficiency through mediating the imbalance between the bone resorptive activity of osteoclasts and the bone formation activity of osteoblasts. Cite this article: W-B. Hsu, W-H. Hsu, J-S. Hung, W-J. Shen, R. W-W. Hsu. Transcriptome analysis of osteoblasts in an ovariectomized mouse model in response to physical exercise. Bone Joint Res 2018;7:601–608. DOI: 10.1302/2046-3758.711.BJR-2018-0075.R2.
Collapse
Affiliation(s)
- W-B Hsu
- Sports Medicine Center, Chang Gung Memorial Hospital, Pu-Tzi City, Taiwan
| | - W-H Hsu
- Sports Medicine Center and Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Pu-Tzi City, Taiwan; Chang Gung University, Pu-Tzi City, Taiwan
| | - J-S Hung
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Pu-Tzi City, Taiwan
| | - W-J Shen
- Po Cheng Orthopedic Institute, Kaohsiung, Taiwan
| | - R W-W Hsu
- Sports Medicine Center and Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Pu-Tzi City, Taiwan; Chang Gung University, Pu-Tzi City, Taiwan
| |
Collapse
|
54
|
Wang Y, Kou J, Zhang H, Wang C, Li H, Ren Y, Zhang Y. The renin-angiotensin system in the synovium promotes periarticular osteopenia in a rat model of collagen-induced arthritis. Int Immunopharmacol 2018; 65:550-558. [PMID: 30412852 DOI: 10.1016/j.intimp.2018.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 01/01/2023]
Abstract
Periarticular osteopenia is the most specific hallmark of rheumatoid arthritis (RA). The renin-angiotensin system (RAS) in the synovium has been found to participate in the pathogenic process of RA. This study examined whether and how RAS regulates periarticular osteopenia in RA. The synovial tissues from patients with RA and osteoarthritis (OA) were prepared. Female Sprague-Dawley rats were treated with either saline, bovine type II collagen (CII) to induce arthritis (CIA), or CII combined with perindopril, an inhibitor of angiotensin-converting enzyme (ACE). Expressions of RAS components, including AT1R, AT2R and ACE, in human and rat synovial tissues were detected. Bone mass of rat joints was examined. Levels of RANKL, OPG and DKK-1 in rat synovium and expressions of TRAF6 and β-catenin in rat bone were examined. The results showed that AT1R, AT2R and ACE in human and rat synovium were up-regulated, but the increased ACE in rat synovial tissues was abrogated by perindopril. While CIA rats displayed increased bone resorption and decreased bone formation, perindopril treatment almost completely abrogated the RAS-mediated osteopenia, indicating that inhibition of ACE reduced the joint damages in rats. The expressions of RANKL and DKK-1 increased in CIA rats as compared with those in the control; TRAF6 was up-regulated and β-catenin was down-regulated in the bone tissues of CIA rats. The changes were then reversed by the use of perindopril. Our findings demonstrate that RAS in the synovium promotes periarticular osteopenia by increasing bone resorption and decreasing bone formation through modulating the RANKL/RANK/TRAF6 and Wnt/β-catenin pathways.
Collapse
Affiliation(s)
- Yingzhen Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, PR China
| | - Jianqiang Kou
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, PR China
| | - Haining Zhang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, PR China
| | - Changyao Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, PR China
| | - Haiyan Li
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, PR China
| | - Yuanzhong Ren
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, PR China
| | - Yongtao Zhang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, PR China.
| |
Collapse
|
55
|
Lazić Mosler E, Lukač N, Flegar D, Fadljević M, Radanović I, Cvija H, Kelava T, Ivčević S, Šućur A, Markotić A, Katavić V, Marušić A, Grčević D, Kovačić N. Fas receptor induces apoptosis of synovial bone and cartilage progenitor populations and promotes bone loss in antigen-induced arthritis. FASEB J 2018; 33:3330-3342. [PMID: 30383451 DOI: 10.1096/fj.201801426r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory joint disease that eventually leads to permanent bone and cartilage destruction. Fas has already been established as the regulator of inflammation in RA, but its role in bone formation under arthritic conditions is not completely defined. The aim of this study was to assess the effect of Fas inactivation on the bone damage during murine antigen-induced arthritis. Subchondral bone of wild-type (WT) and Fas-knockout (Fas-/-) mice was evaluated by histomorphometry and microcomputerized tomography. Proportions of synovial bone and cartilage progenitors were assessed by flow cytometry. Synovial bone and cartilage progenitors were purified by fluorescence-activated cell sorting and expression of Fas and Fas-induced apoptosis were analyzed in vitro. Results showed that Fas-/- mice developed attenuated arthritis characterized by preserved epiphyseal bone and cartilage. A proportion of the earliest CD200+ bone and cartilage progenitors was reduced in WT mice with arthritis and was unaltered in Fas-/- mice. During osteoblastic differentiation in vitro, CD200+ cells express the highest levels of Fas and are removed by Fas ligation. These results suggest that Fas-induced apoptosis of early CD200+ osteoprogenitor population represents potential mechanism underlying the impaired bone formation in arthritis, so their preservation may represent the bone-protective mechanism during arthritis.-Lazić Mosler, E., Lukač, N., Flegar, D., Fadljević, M., Radanović, I., Cvija, H., Kelava, T., Ivčević, S., Šućur, A., Markotić, A., Katavić, V., Marušić, A., Grčević, D., Kovačić, N. Fas receptor induces apoptosis of synovial bone and cartilage progenitor populations and promotes bone loss in antigen-induced arthritis.
Collapse
Affiliation(s)
- Elvira Lazić Mosler
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Dermatology and Venerology, General Hospital Dr. Ivo Pedišić, Sisak, Croatia.,Department of Nursing, Catholic University of Croatia, Zagreb, Croatia
| | - Nina Lukač
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Darja Flegar
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Martina Fadljević
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Igor Radanović
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Hrvoje Cvija
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tomislav Kelava
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sanja Ivčević
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Alan Šućur
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Antonio Markotić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Centre for Clinical Pharmacology, University Clinical Hospital Mostar, Mostar, Bosnia and Herzegovina; and
| | - Vedran Katavić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Marušić
- Department of Research in Biomedicine and Health, University of Split School of Medicine, Split, Croatia
| | - Danka Grčević
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
56
|
Pan YJ, Wang WH, Huang TY, Weng WH, Fang CK, Chen YC, Hwang JJ. Quetiapine ameliorates collagen-induced arthritis in mice via the suppression of the AKT and ERK signaling pathways. Inflamm Res 2018; 67:847-861. [PMID: 30109356 DOI: 10.1007/s00011-018-1176-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 06/12/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE AND DESIGN To investigate the amelioration effects of quetiapine on rheumatoid arthritis with RAW 264.7 macrophage and collagen-induced arthritis (CIA) DBA/1J mouse model. SUBJECTS RAW 264.7 macrophage and DBA/1J mice. TREATMENT Lipopolysaccharide and collagen. METHODS RAW 264.7 macrophages stimulated by lipopolysaccharide (LPS) followed by quetiapine treatments were investigated. Activations of CD80 and CD86 were analyzed by flow cytometry. Pro-inflammatory cytokines such as IL-6, TNF-α and IL-1β were analyzed by ELISA. Proteins involved in signaling pathways related to the formation of rheumatoid arthritis were assayed by Western blotting. Therapeutic efficacy of quetiapine in CIA mouse model was also assayed. 18F-FDG/micro-PET was used to monitor the inflammation status in the joints, and the severity of bone erosion was evaluated with micro-CT and H&E staining. RESULTS The inhibition of pro-inflammatory cytokines by quetiapine was found through the ERK and AKT phosphorylation and subsequent NF-κB and CREB signaling pathways. Pro-inflammatory cytokines such as IL-17, IL-6 and IL-1β were decreased, while immunosuppressive factors such as TGF-β and IL-10 were increased in CIA mice treated with quetiapine. Notably, no uptake of 18F-FDG and bone erosion was found with micro-PET images on days 32 and 43 in the quetiapine-treated and normal control groups. However, significant uptake of 18F-FDG could be observed in the CIA group during the same time course. Similar results were further verified with ex vivo autoradiography. CONCLUSION Taken together, these results suggest that quetiapine is a potential anti-inflammatory drug, and may be used as an adjuvant for the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Yi-Ju Pan
- Department of Psychiatry, Far Eastern Memorial Hospital, Banciao, New Taipei City, 220, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Hsun Wang
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan.,Department of Medical Imaging and Radiology, Shu-Zen Junior College of Medicine and Management, Kao-hsiung, Taiwan
| | - Tzu-Yao Huang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Hsiang Weng
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Kai Fang
- Department of Psychiatry and Suicide Prevention Center, Mackay Memorial Hospital, No. 155, Sec.2, Li-Nong Street, Bei-tou, Taipei, 112, Taiwan
| | - Yu-Chan Chen
- Department of Radiation Oncology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Jeng-Jong Hwang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan. .,Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
57
|
Tang M, Tian L, Luo G, Yu X. Interferon-Gamma-Mediated Osteoimmunology. Front Immunol 2018; 9:1508. [PMID: 30008722 PMCID: PMC6033972 DOI: 10.3389/fimmu.2018.01508] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/18/2018] [Indexed: 02/05/2023] Open
Abstract
Osteoimmunology is the interdiscipline that focuses on the relationship between the skeletal and immune systems. They are interconnected by shared signal pathways and cytokines. Interferon-gamma (IFN-γ) plays important roles in immune responses and bone metabolism. IFN-γ enhances macrophage activation and antigen presentation. It regulates antiviral and antibacterial immunity as well as signal transduction. IFN-γ can promote osteoblast differentiation and inhibit bone marrow adipocyte formation. IFN-γ plays dual role in osteoclasts depending on its stage. Furthermore, IFN-γ is an important pathogenetic factor in some immune-mediated bone diseases including rheumatoid arthritis, postmenopausal osteoporosis, and acquired immunodeficiency syndrome. This review will discuss the contradictory findings of IFN-γ in osteoimmunology and its clinical application potential.
Collapse
Affiliation(s)
- Mengjia Tang
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Tian
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guojing Luo
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
58
|
Garrido-Castro JL, Curbelo R, Mazzucchelli R, Domínguez-González ME, Gonzalez-Navas C, Flores Robles BJ, Zarco P, Mulero J, Cea-Calvo L, Arteaga MJ, Font-Ugalde P, Carmona L, Collantes-Estevez E. High Reproducibility of an Automated Measurement of Mobility for Patients with Axial Spondyloarthritis. J Rheumatol 2018; 45:1383-1388. [PMID: 29907675 DOI: 10.3899/jrheum.170941] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Conventional measures of spinal mobility used in the assessment of patients with axial spondyloarthritis (axSpA), such as the Bath Ankylosing Spondylitis Metrology Index and its components, are subject to interobserver variability. The University of Córdoba Ankylosing Spondylitis Metrology Index (UCOASMI) is a validated composite index based on a motion video-capture system, UCOTrack. Our objective was to assess its reproducibility in clinical practice settings. METHODS We carried out an observational study of repeated measures in 3 centers. Video-capture systems were installed and adapted to clinical rooms. Patients with axSpA and stable disease were selected by consecutive stratified sampling [disease duration, sex, and the Bath Ankylosing Spondylitis Disease Activity Index (BASDAI)]. Intraobserver reliability of the UCOASMI and of conventional measures was tested 3-5 days apart. For interobserver reliability, 3 patients from each center were evaluated in the other centers, within 3-7 days. The intraclass correlation coefficients (ICC) were calculated. RESULTS Thirty patients were included (73% men, mean age 53 yrs, mean BASDAI 3.0). Interobserver and intraobserver ICC of the UCOASMI was 0.98. Conventional measurements showed lower but adequate reproducibility as well, except for interobserver reliability of lateral flexion (0.41), cervical rotation (0.61), and Schöber test (0.07), and intraobserver reliability of tragus-to-wall distance (0.30). CONCLUSION Reproducibility of the UCOASMI seems very high, and apparently more reliable than conventional measures of mobility.
Collapse
Affiliation(s)
- Juan L Garrido-Castro
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain. .,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba.
| | - Rafael Curbelo
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Ramón Mazzucchelli
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - María E Domínguez-González
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Cristina Gonzalez-Navas
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Bryan J Flores Robles
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Pedro Zarco
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Juan Mulero
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Luis Cea-Calvo
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - María J Arteaga
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Pilar Font-Ugalde
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Loreto Carmona
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| | - Eduardo Collantes-Estevez
- From the Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC); Department of Rheumatology, Hospital Universitario Reina Sofía; Department of Medicine, Universidad de Córdoba, Córdoba; Instituto de Salud Musculoesquelética; Department of Rheumatology, Hospital Fundación Alcorcón, Alcorcón; Department of Rheumatology, Hospital Puerta de Hierro, Majadahonda; Medical Affairs, Merck Sharp & Dohme, Madrid, Spain.,J.L. Garrido-Castro, IE, PhD, IMIBIC; R. Curbelo, PT, PhD, Instituto de Salud Musculoesquelética; R. Mazzucchelli, MD, Department of Rheumatology, Hospital Fundación Alcorcón; M.E. Domínguez-González, RN, Department of Rheumatology, Hospital Fundación Alcorcón; C. Gonzalez-Navas, RN, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía; B.J. Flores Robles, MD, Department of Rheumatology, Hospital Puerta de Hierro; P. Zarco, MD, PhD, Department of Rheumatology, Hospital Fundación Alcorcón; J. Mulero, MD, PhD, Department of Rheumatology, Hospital Puerta de Hierro; L. Cea-Calvo, MD, Medical Affairs, Merck Sharp & Dohme; M.J. Arteaga, MD, Medical Affairs, Merck Sharp & Dohme; P. Font-Ugalde, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba; L. Carmona, MD, PhD, Instituto de Salud Musculoesquelética; E. Collantes-Estevez, MD, PhD, IMIBIC, and Department of Rheumatology, Hospital Universitario Reina Sofía, and Department of Medicine, Universidad de Córdoba
| |
Collapse
|
59
|
Ohgi K, Kajiya H, Goto-T K, Okamoto F, Yoshinaga Y, Okabe K, Sakagami R. Toll-like receptor 2 activation primes and upregulates osteoclastogenesis via lox-1. Lipids Health Dis 2018; 17:132. [PMID: 29859535 PMCID: PMC5985062 DOI: 10.1186/s12944-018-0787-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 05/23/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lectin-like oxidized low-density-lipoprotein receptor 1 (Lox-1) is the receptor for oxidized low-density lipoprotein (oxLDL), a mediator in dyslipidemia. Toll-like receptor (TLR)-2 and - 4 are receptors of lipopolysaccharide (LPS) from Porphyromonas gingivalis, a major pathogen of chronic periodontitis. Although some reports have demonstrated that periodontitis has an adverse effect on dyslipidemia, little is clear that the mechanism is explained the effects of dyslipidemia on osteoclastogenesis. We have hypothesized that osteoclast oxLDL has directly effect on osteoclasts (OCs), and therefore alveolar bone loss on periodontitis may be increased by dyslipidemia. The present study aimed to elucidate the effect of Lox-1 on osteoclastogenesis associated with TLRs in vitro. METHODS Mouse bone marrow cells (BMCs) were stimulated with macrophage colony-stimulating factor into bone marrow macrophages (BMMs). The cells were also stimulated with synthetic ligands for TLR2 (Pam3CSK4) or TLR4 (Lipid A), with or without receptor activator of nuclear factor kappa-B ligand (RANKL), and assessed for osteoclastogenesis by tartrate-resistant acid phosphatase (TRAP) staining, immunostaining, western blotting, flow activated cell sorting (FACS) analysis, real-time polymerase chain reaction (PCR), and reverse transcription PCR. RESULTS Lox-1 expression was significantly upregulated by Pam3CSK4 and Lipid A in BMCs (p < 0.05), but not in BMMs. FACS analysis identified that Pam3CSK4 upregulated RANK and Lox-1 expression in BMCs. TRAP-positive cells were not increased by stimulation with Pam3CSK4 alone, but were increased by stimulation with combination combined Pam3CSK and oxLDL. Expression of both Lox-1 and myeloid differentiation factor 88 (MyD88), an essential adaptor protein in the TLR signaling pathway, were suppressed by inhibitors of TLR2, TLR4 and mitogen-activated protein kinase (MAPK). CONCLUSIONS This study supports that osteoclastogenesis is promoted under the coexistence of oxLDL by TLR2-induced upregulation of Lox-1 in BMCs. This indicates that periodontitis could worsen with progression of dyslipidemia.
Collapse
Affiliation(s)
- Kimiko Ohgi
- Department of Odontology, Fukuoka Dental College, Fukuoka, 8140193, Japan
| | - Hiroshi Kajiya
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, 8140193, Japan.
| | - Kazuko Goto-T
- Department of Dental Hygiene, Fukuoka College of Health Sciences, Fukuoka, 8140193, Japan
| | - Fujio Okamoto
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, 8140193, Japan
| | - Yasunori Yoshinaga
- Department of Odontology, Fukuoka Dental College, Fukuoka, 8140193, Japan
| | - Koji Okabe
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, 8140193, Japan
| | - Ryuji Sakagami
- Department of Odontology, Fukuoka Dental College, Fukuoka, 8140193, Japan
| |
Collapse
|
60
|
Ahmed HS, Farrag SE, Okasha AE, Othman G, Shady I. Risk Factors for Systemic Reduced Bone Mineral Density in
Premenopausal Female Patients with Early Untreated Rheumatoid
Arthritis. AKTUEL RHEUMATOL 2018. [DOI: 10.1055/a-0591-2364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Abstract
Background Systemic osteoporosis (OP) is evident among patients with
early rheumatoid arthritis (ERA). This study aimed to investigate the OP risk
factors in patients with ERA and who was treatment-naïve at
inclusion.
Subjects and Methods Systemic bone mineral density (BMD) of the lumbar
spine (LS), femoral neck (FN) and total hip (TH) was measured in 135
treatment-naïve premenopausal females with early Rheumatoid Arthritis
(ERA). For all patients, demographic data, vitamin D status, and the specific
parameters of the disease, including disease activity, serum levels of
rheumatoid factor and anti-citrullinated protein antibodies (ACPA) were
evaluated.
Results T score was<−1.0 in the LS in 16.2%, in
the FN in 22.2% and in the TH in 23.7%. Among our patients,
29.6% had below normal T score at any site. Demographic characteristics,
RA duration, diseases activity did not significantly impact BMD. However,
patients with decreased BMD were more prevalent ACPA- and rheumatoid factor
(RF)-positive than patients with normal BMD. Also, high titer ACPA or RF is
associated with more marked reduction in BMD. In regression analysis, after
adjustment for possible confounders, patient stratification according to ACPA
status and RF status (into negative, low-positive and high positive) still a
significant independent variable associated with lower BMD values.
Conclusion Presence of ACPA or RF is associated with increased risk for
development of reduced systemic BMD from very early stage of rheumatoid
arthritis. Furthermore, this risk increases more with higher levels of ACPA or
RF. Measurement of BMD should be performed for ACPA- or RF-positive patients
with early RA.
Collapse
Affiliation(s)
- Hamada S. Ahmed
- Mansoura University Faculty of Medicine, Rheumatology and Reahb. Dep,
Mansoura, Egypt
| | - Sherif E. Farrag
- Mansoura University Faculty of Medicine, Rheumatology and Reahb. Dep,
Mansoura, Egypt
| | - Amr E. Okasha
- Mansoura University Faculty of Medicine, Rheumatology and Reahb. Dep,
Mansoura, Egypt
| | - Gamal Othman
- Mansoura University Faculty of Medicine, Biochemistry Dep., Mansoura,
Egypt
| | - Ibrahim Shady
- Mansoura University Faculty of Medicine, Public Health and community
medicine, Mansoura, Egypt
| |
Collapse
|
61
|
Ibrahim-Nasser N, Marotte H, Valery A, Salliot C, Toumi H, Lespessailles E. Precision and sources of variability in the assessment of rheumatoid arthritis erosions by HRpQCT. Joint Bone Spine 2018; 85:211-217. [DOI: 10.1016/j.jbspin.2017.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 02/15/2017] [Indexed: 01/14/2023]
|
62
|
Ahmad HA, Alemao E, Guo Z, Iannaccone CK, Frits ML, Weinblatt M, Shadick NA. Association of Low Bone Mineral Density with Anti-Citrullinated Protein Antibody Positivity and Disease Activity in Established Rheumatoid Arthritis: Findings from a US Observational Cohort. Adv Ther 2018; 35:232-242. [PMID: 29368271 PMCID: PMC5818577 DOI: 10.1007/s12325-017-0657-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Indexed: 01/01/2023]
Abstract
Introduction To assess the relationship between low bone mineral density (BMD), anti-cyclic citrullinated peptide-2 (anti-CCP2) antibodies, and disease activity in patients with established rheumatoid arthritis (RA). Methods Patients enrolled in a single-center, observational cohort registry of patients with RA. Eligible patients had known BMD, as measured by digital X-ray radiogrammetry (DXR–BMD), and anti-CCP2 antibody measurements at the same time point or within 6 months. Anti-CCP2–immunoglobulin (Ig)G-positive (+) patients (≥ 20 U/mL) were distributed into three equal groups (Gp1–3), representing increasing anti-CCP2 antibody concentrations. Associations between BMD and anti-CCP2 antibody status and titer were explored in multivariate regression analyses controlling for covariates (including age, duration of RA, use of steroids, use of osteoporosis medication). Association between disease activity (DAS28 [CRP] < 2.6) and bone loss was also explored. Results A total of 149 patients (all women) were included (47 anti-CCP2 antibody negative [−], 102 anti-CCP2+ [34\titer group]). Mean disease duration was greater in the three anti-CCP2+ groups vs. the anti-CCP2− group. DXR–BMD was lower in the anti-CCP2+ vs. the anti-CCP2− groups (Gp1–3 vs. anti-CCP2−: P < 0.0001 for left and right hands). DXR–BMD decreased with increasing anti-CCP2 titer (P < 0.001 for left and right hands). Patients with low DXR–BMD were less likely to have a DAS28 (CRP) < 2.6 (P = 0.0181). Conclusion Among patients with established RA, data suggest that anti-CCP2+ patients, particularly those with high anti-CCP2 antibody titers, have lower hand BMD, and patients with lower hand BMD are less likely to have low disease activity. Funding Bristol-Myers Squibb. Trial Registration Clinicaltrials.gov identifier, NCT01793103. Electronic supplementary material The online version of this article (10.1007/s12325-017-0657-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Evo Alemao
- Bristol-Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | |
Collapse
|
63
|
Kittaka M, Mayahara K, Mukai T, Yoshimoto T, Yoshitaka T, Gorski JP, Ueki Y. Cherubism Mice Also Deficient in c-Fos Exhibit Inflammatory Bone Destruction Executed by Macrophages That Express MMP14 Despite the Absence of TRAP+ Osteoclasts. J Bone Miner Res 2018; 33:167-181. [PMID: 28914985 PMCID: PMC5771992 DOI: 10.1002/jbmr.3295] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/28/2017] [Accepted: 09/07/2017] [Indexed: 11/10/2022]
Abstract
Currently, it is believed that osteoclasts positive for tartrate-resistant acid phosphatase (TRAP+) are the exclusive bone-resorbing cells responsible for focal bone destruction in inflammatory arthritis. Recently, a mouse model of cherubism (Sh3bp2KI/KI ) with a homozygous gain-of-function mutation in the SH3-domain binding protein 2 (SH3BP2) was shown to develop auto-inflammatory joint destruction. Here, we demonstrate that Sh3bp2KI/KI mice also deficient in the FBJ osteosarcoma oncogene (c-Fos) still exhibit noticeable bone erosion at the distal tibia even in the absence of osteoclasts at 12 weeks old. Levels of serum collagen I C-terminal telopeptide (ICTP), a marker of bone resorption generated by matrix metalloproteinases (MMPs), were elevated, whereas levels of serum cross-linked C-telopeptide (CTX), another resorption marker produced by cathepsin K, were not increased. Collagenolytic MMP levels were increased in the inflamed joints of the Sh3bp2KI/KI mice deficient in c-Fos. Resorption pits contained a large number of F4/80+ macrophages and genetic depletion of macrophages rescued these erosive changes. Importantly, administration of NSC405020, an MMP14 inhibitor targeted to the hemopexin (PEX) domain, suppressed bone erosion in c-Fos-deficient Sh3bp2KI/KI mice. After activation of the NF-κB pathway, macrophage colony-stimulating factor (M-CSF)-dependent macrophages from c-Fos-deficient Sh3bp2KI/KI mice expressed increased amounts of MMP14 compared with wild-type macrophages. Interestingly, receptor activator of NF-κB ligand (RANKL)-deficient Sh3bp2KI/KI mice failed to show notable bone erosion, whereas c-Fos deletion did restore bone erosion to the RANKL-deficient Sh3bp2KI/KI mice, suggesting that osteolytic transformation of macrophages requires both loss-of-function of c-Fos and gain-of-function of SH3BP2 in this model. These data provide the first genetic evidence that cells other than osteoclasts can cause focal bone destruction in inflammatory bone disease and suggest that MMP14 is a key mediator conferring pathological bone-resorbing capacity on c-Fos-deficient Sh3bp2KI/KI macrophages. In summary, the paradigm that osteoclasts are the exclusive cells executing inflammatory bone destruction may need to be reevaluated based on our findings with c-Fos-deficient cherubism mice lacking osteoclasts. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mizuho Kittaka
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kotoe Mayahara
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,Department of Orthodontics, Nihon University, School of Dentistry, Tokyo, Japan
| | - Tomoyuki Mukai
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Tetsuya Yoshimoto
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Teruhito Yoshitaka
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Jeffrey P Gorski
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,University of Missouri-Kansas City (UMKC) Center of Excellence in the Study of Dental and Musculoskeletal Tissues (CEMT), Kansas City, MO, USA
| | - Yasuyoshi Ueki
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA.,University of Missouri-Kansas City (UMKC) Center of Excellence in the Study of Dental and Musculoskeletal Tissues (CEMT), Kansas City, MO, USA
| |
Collapse
|
64
|
Yu H, Jiang L, Wan B, Zhang W, Yao L, Che T, Gan C, Su N, He J, Huang J, Zhang K, Zhang Y. The role of aryl hydrocarbon receptor in bone remodeling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 134:44-49. [PMID: 29277341 DOI: 10.1016/j.pbiomolbio.2017.12.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022]
Abstract
Bone remodeling is a persistent process for maintaining skeletal system homeostasis, and it depends on the dynamic equilibrium between bone-forming osteoblasts and bone-resorbing osteoclasts. Aryl hydrocarbon receptor (Ahr), a ligand-activated transcription factor, plays a pivotal role in regulating skeletal system. In order to better understand the role of Ahr in bone remodeling, we focused on bone remodeling characteristic, and the effects of Ahr on bone formation and differentiation, which suggest that Ahr is a critical control factor in the process of bone remodeling. Moreover, we discussed the impacts of Ahr on several signaling pathways related to bone remodeling, hoping to provide a theoretical basis to improve bone remodeling.
Collapse
Affiliation(s)
- Haitao Yu
- Department of Clincal Laboratory, The First Hospital of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China; The First Clinical College of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China.
| | - Lili Jiang
- School of Material Science and Technology, Lanzhou University of Technology, Langongping Road, Lanzhou 730050, Gansu Province, PR China
| | - Bo Wan
- The 3rd and 4th Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Wei Zhang
- Cental Laboratory, The First Hospital of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Liqiong Yao
- Department of Clincal Laboratory, The First Hospital of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Tuanjie Che
- Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, East road no. 110 nanhe yantan, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Chao Gan
- Department of Clincal Laboratory, The First Hospital of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Na Su
- Department of Clincal Laboratory, The First Hospital of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Jinchun He
- Department of Clincal Laboratory, The First Hospital of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Jintian Huang
- The First Clinical College of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Kaiyun Zhang
- The First Clinical College of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| | - Yiheng Zhang
- The First Clinical College of Lanzhou University, West Road No. 1 East Hills, Chengguan District, Lanzhou, 730000, Gansu Province, PR China
| |
Collapse
|
65
|
Tan X, Huang D, Zhou W, Yan L, Yue J, Lu W, Song D, Zhou X, Ye L, Zhang L. Dickkopf-1 may regulate bone coupling by attenuating wnt/β-catenin signaling in chronic apical periodontitis. Arch Oral Biol 2017; 86:94-100. [PMID: 29216526 DOI: 10.1016/j.archoralbio.2017.11.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/20/2017] [Accepted: 11/27/2017] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Alveolar bone loss is a common outcome of chronic apical periodontitis. In this study, we investigated the involvement of the Dickkopf-1-Wnt/β-catenin signaling pathway in the attenuation of osteogenic differentiation induced by Escherichia coli lipopolysaccharide, and we evaluated the use of Dickkopf-1 inhibitor and Dickkopf-1 recombinant protein to reverse bone loss in different phases of osteogenic differentiation. METHODS MC3T3-E1 cells grown in osteogenic medium were treated with Escherichia coli lipopolysaccharide for 24h during osteogenic induction on days 0, 1, 7, 14 and 21. Dickkopf-1 siRNA was added on days 0 and 1, and Dickkopf-1 recombinant was added on days 7, 14, and 21. Quantitative real-time PCR, Western blotting and alkaline phosphatase activity assays were performed to measure osteogenic marker expression and Wnt/β-catenin signaling. A rat apical periodontitis model was used to further evaluate the function of Dickkopf-1 in relation to bone loss. RESULTS MC3T3-E1 cells treated with Escherichia coli lipopolysaccharide showed decreased mRNA expression of osteogenic markers. Wnt/β-catenin signaling was also inhibited, and Dickkopf-1 showed corresponding variations as quantified by Western blotting. Using Dickkopf-1 inhibitor or Dickkopf-1 recombinant protein at different phases of osteogenic differentiation in vitro partially reversed the decrease in osteogenic marker expression. The rat apical periodontitis model indicated that the Dickkopf-1 inhibitor could restore bone loss in the periapical area in vivo. CONCLUSIONS Dickkopf-1 may play a key regulatory role in determining the outcome for bone in inflammatory environments, and modulating the Wnt/β-catenin signaling pathway via Dickkopf-1 inhibitor or recombinant protein may provide a potential therapeutic option to prevent bone destruction in endodontic disease.
Collapse
Affiliation(s)
- Xuelian Tan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China.
| | - Wei Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Li Yan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Junli Yue
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - WanLu Lu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University, 14# 3rd Section, Renmin South Road, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
66
|
Sapir-Koren R, Livshits G. Postmenopausal osteoporosis in rheumatoid arthritis: The estrogen deficiency-immune mechanisms link. Bone 2017; 103:102-115. [PMID: 28666971 DOI: 10.1016/j.bone.2017.06.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/13/2017] [Accepted: 06/26/2017] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis (RA) is characterized, among other factors, by systemic bone loss, reaching ~50% prevalence of osteoporosis in postmenopausal women. This is roughly a doubled prevalence in comparison with age-matched non-RA women. Postmenopausal RA women are more likely to be sero-positive for the anti-citrullinated peptide antibody (ACPA). Our extensive review of recent scientific literature enabled us to propose several mechanisms as responsible for the accelerated bone loss in ACPA(+) RA postmenopausal women. Menopause-associated estrogen deficiency plays a major role in these pathological mechanisms, as follows.
Collapse
Affiliation(s)
- Rony Sapir-Koren
- Human Population Biology Research Group, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Gregory Livshits
- Human Population Biology Research Group, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Lilian and Marcel Pollak Chair of Biological Anthropology, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
67
|
Possible role of Dickkopf-1 protein in the pathogenesis of tympanosclerosis in a rat model. The Journal of Laryngology & Otology 2017; 131:860-865. [PMID: 28807065 DOI: 10.1017/s0022215117001566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES This study aimed to investigate the expression of DKK1 protein in an experimental model of tympanosclerosis and its possible role in the pathogenesis of this disorder. METHODS Forty Sprague Dawley rats were included in the study: 20 in the control group (which received no treatment) and 20 in the experimental group (which received an incision to induce tympanosclerosis). Otomicroscopy was performed to observe the development of myringosclerosis. Haematoxylin and eosin staining was performed to observe the morphological changes. Western blot analysis and immunohistochemistry were performed to assess the expression of DKK1 protein. RESULTS At day 15, sclerotic lesions were observed in 70 per cent of the tympanic membranes. Inflammatory infiltration and hyaline degeneration markedly appeared in the tympanic membranes and middle-ear mucosa. DKK1 protein was mainly distributed in the cytoplasm of epithelial cells, which were widely distributed in the tympanic membranes and middle-ear mucosa. The expression of DKK1 protein was significantly decreased in the calcified experimental ears. CONCLUSION DKK1 protein is involved in the pathogenesis of tympanosclerosis by regulating the Wnt/β-catenin signalling pathway.
Collapse
|
68
|
Cellular and molecular pathways of structural damage in rheumatoid arthritis. Semin Immunopathol 2017; 39:355-363. [PMID: 28597065 DOI: 10.1007/s00281-017-0634-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/19/2017] [Indexed: 12/11/2022]
Abstract
Structural damage of cartilage and bone tissue is a hallmark of rheumatoid arthritis (RA). The resulting joint destruction constitutes one of the major disease consequences for patients and creates a significant burden for the society. The main cells executing bone and cartilage degradation are osteoclasts and fibroblast-like synoviocytes, respectively. The function of both cell types is heavily influenced by the immune system. In the last decades, research has identified several mediators of structural damage, ranging from infiltrating immune cells and inflammatory cytokines to autoantibodies. These factors result in an inflammatory milieu in the affected joints which leads to an increased development and function of osteoclasts and the transformation of fibroblast-like synoviocytes towards a highly migratory and destructive phenotype. In addition, repair mechanisms mediated by osteoblasts and chondrocytes are strongly impaired by the presence of pro-inflammatory cytokines. This article will review the current knowledge on the mechanisms of joint inflammation and the destruction of bone and cartilage.
Collapse
|
69
|
Andrade KMD, Alfenas BFM, Campos CH, Rodrigues Garcia RCM. Mandibular movements in older people with rheumatoid arthritis. Oral Surg Oral Med Oral Pathol Oral Radiol 2017; 123:e153-e159. [PMID: 28407990 DOI: 10.1016/j.oooo.2017.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The aim of this study was to compare the mandibular movements in older people with and without temporomandibular disorder (TMD) associated with rheumatoid arthritis (RA). STUDY DESIGN Thirty partially or completely edentulous older adults (65.33 ± 4.7 years) were assigned to 2 groups: (1) with RA and TMD and (2) without RA and TMD. Chewing movements of the jaws during mastication of the test material (Optocal) and the range of mandibular movements were evaluated by using the JT-3-D kinesiographic device before and after new removable prosthesis insertion. Multiple comparisons were made with analysis of variance (ANOVA) and the Tukey-Kramer test. RESULTS Comparisons between the 2 groups before and after new prosthesis insertion revealed that the RA and TMD group had reduced opening angles (P < .05) during chewing. After insertion of new prostheses, both groups showed increased opening and closing angles during chewing (P < .05). The mandibular range of motion results showed that patients with RA and TMD exhibited lower aperture and laterality movements (P < .05) compared with controls before and after new prosthesis insertion. However, there was an increase in aperture, lefty laterality, and protrusion values after new prosthesis insertion in both groups. CONCLUSIONS TMD associated with RA may impair mandibular movements. Well-fitted prostheses may improve mandibular movements in older adults, especially those with RA.
Collapse
Affiliation(s)
- Kelly Machado de Andrade
- Graduate Student, Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Bruna Fernandes Moreira Alfenas
- Graduate Student, Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Camila Heitor Campos
- Assistant Professor, Department of Specific Formation, Nova Friburgo Health Institute, Fluminense Federal University, Rio de Janeiro, Brazil
| | | |
Collapse
|
70
|
Liu J, Ren X, Zhang M, Lei Y, Chen Y, He H. Roles of Wnt3a and Dkk1 in experimental periodontitis. J Dent Sci 2017; 12:220-225. [PMID: 30895054 PMCID: PMC6400003 DOI: 10.1016/j.jds.2016.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 12/20/2022] Open
Abstract
Background/purpose Periodontitis is an inflammatory, destructive disease caused by periodontal bacteria, and its molecular mechanism remains unclear. The aims of this study are to evaluate the expressions of Wnt3a and Dkk1 in experimental periodontitis (EP) and preliminarily explore their roles in periodontal diseases. Materials and methods A total of 64 six-week-old male Sprague–Dawley rats were randomly divided into a normal group and an EP group. The EP group was prepared by using silk ligature combined with intraoral bacteria inoculation. To assess the periodontal inflammation and bone destruction extent, hematoxylin and eosin staining and tartrate-resistant acid phosphatase staining was performed 2 weeks, 4 weeks, and 6 weeks after the modeling, respectively, and immunohistochemistry and enzyme-linked immunosorbent assay were also performed to detect the changes of Wnt3a and Dkk1 in periodontal tissue and plasma. Results Wnt3a expression was significantly decreased in the EP group when compared with the normal group (P < 0.05). Meanwhile, Dkk1 expression was significantly increased in the EP group when compared with the normal group (P < 0.05). Conclusion The expression of Wnt3a and Dkk1 was well correlated with EP. It is suggested that Wnt3a and Dkk1 may be involved in periodontal diseases.
Collapse
Affiliation(s)
- Jianqi Liu
- Department of Dentistry, the Affiliated Yan'an Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiaobin Ren
- Department of Periodontology, the Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Mingzhu Zhang
- Department of Periodontology, the Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yayan Lei
- Department of Endodontology, the Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuhua Chen
- Department of Dentistry, Wuxi Mental Health Center, Jiang Su, China
| | - Hongbing He
- Department of Periodontology, the Affiliated Stomatological Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
71
|
Boman A, Kokkonen H, Ärlestig L, Berglin E, Rantapää-Dahlqvist S. Receptor activator of nuclear factor kappa-B ligand (RANKL) but not sclerostin or gene polymorphisms is related to joint destruction in early rheumatoid arthritis. Clin Rheumatol 2017; 36:1005-1012. [PMID: 28190118 PMCID: PMC5400786 DOI: 10.1007/s10067-017-3570-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/22/2017] [Accepted: 01/31/2017] [Indexed: 01/25/2023]
Abstract
The aim of this study was to analyze relationships between receptor activator of nuclear factor kappa-B (RANKL), sclerostin and their gene polymorphisms with radiological progression in patients with early rheumatoid arthritis (RA). Patients with early RA (n = 407, symptomatic <1 year) (ARA criteria) examined radiologically at inclusion and after 24 months were consecutively included. Disease activity score and C-reactive protein were regularly recorded. Sclerostin, RANKL, and anti-CCP2 antibodies were analyzed in plasma at baseline using ELISAs. Data on gene polymorphism for sclerostin and RANKL were extracted from Immunochip analysis. Sex- and age-matched controls (n = 71) were identified from the Medical Biobank of Northern Sweden. The concentration of RANKL was significantly higher in patients compared with controls, median (IQR) 0.56 (0.9) nmol/L and 0.20 (0.25) nmol/L (p < 0.001), and in anti-CCP2-positive patients compared with sero-negative individuals. Sclerostin was significantly increased in female patients 0.59 (0.47–0.65) ng/mL compared with female controls 0.49 (0.4–0.65) ng/mL (p < 0.02). RANKL concentration was related to the Larsen score at baseline (p < 0.01), after 24 months (p < 0.001), and to radiological progression at 24 months (p < 0.001). Positivity of RANKL and anti-CCP2 yielded significant risk for progression with negativity for both as reference. No single nucleotide polymorphism encoding TNFSF11 or SOST was associated with increased concentrations of the factors. The concentration of RANKL was related to the Larsen score at baseline, at 24 months, and radiological progression at 24 months particularly in anti-CCP2-positive patients, while the concentration of sclerostin was unrelated to radiological findings.
Collapse
Affiliation(s)
- Antonia Boman
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | - Heidi Kokkonen
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | - Lisbeth Ärlestig
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | - Ewa Berglin
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | | |
Collapse
|
72
|
Baum R, Gravallese EM. Bone as a Target Organ in Rheumatic Disease: Impact on Osteoclasts and Osteoblasts. Clin Rev Allergy Immunol 2017; 51:1-15. [PMID: 26411424 DOI: 10.1007/s12016-015-8515-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dysregulated bone remodeling occurs when there is an imbalance between bone resorption and bone formation. In rheumatic diseases, including rheumatoid arthritis (RA) and seronegative spondyloarthritis, systemic and local factors disrupt the process of physiologic bone remodeling. Depending upon the local microenvironment, cell types, and local mechanical forces, inflammation results in very different effects on bone, promoting bone loss in the joints and in periarticular and systemic bone in RA and driving bone formation at enthesial and periosteal sites in diseases such as ankylosing spondylitis (AS), included within the classification of axial spondyloarthritis. There has been a great deal of interest in the role of osteoclasts in these processes and much has been learned over the past decade about osteoclast differentiation and function. It is now appreciated that osteoblast-mediated bone formation is also inhibited in the RA joint, limiting the repair of erosions. In contrast, osteoblasts function to produce new bone in AS. The Wnt and BMP signaling pathways have emerged as critical in the regulation of osteoblast function and the outcome for bone in rheumatic diseases, and these pathways have been implicated in both bone loss in RA and bone formation in AS. These pathways provide potential novel approaches for therapeutic intervention in diseases in which inflammation impacts bone.
Collapse
Affiliation(s)
- Rebecca Baum
- Department of Medicine and Division of Rheumatology, University of Massachusetts Medical School, Lazare Research Building Suite 223, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Ellen M Gravallese
- Department of Medicine and Division of Rheumatology, University of Massachusetts Medical School, Lazare Research Building Suite 223, 364 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
73
|
Sousa LHT, Moura EV, Queiroz AL, Val D, Chaves H, Lisboa M, Furlaneto F, Brito GA, Goes P. Effects of glucocorticoid-induced osteoporosis on bone tissue of rats with experimental periodontitis. Arch Oral Biol 2017; 77:55-61. [PMID: 28178585 DOI: 10.1016/j.archoralbio.2017.01.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 01/04/2017] [Accepted: 01/17/2017] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To evaluate the effects of osteoporosis induced by glucocorticoid (GIOP) on bone tissue of rats with experimental periodontitis (EP). DESIGN 48 male Wistar rats divided into groups: Naïve, EP, GIOP and GIOP+EP. Rats of GIOP and GIOP+EP groups received 7mg/kg of dexamethasone intramuscularly once a week for 5 weeks. Following, EP and GIOP+EP groups were subjected to ligature-induced periodontitis. Naïve group experienced no manipulation. After 11 days, the animals were euthanized and left maxillae collected for macroscopic, radiographic, micro-tomographic and microscopic analysis of alveolar bone loss (ABL). Blood samples were collected for determination of bone-specific alkaline phosphatase (BALP) levels and the right femurs were removed for radiographic and biomechanical analysis. RESULTS EP caused ABL and reduced BALP levels (p<0,05), but it did not change the architecture or biomechanics of femur, compared to Naïve. GIOP did not cause ABL, but it significantly decreased alveolar bone mineral density (ABMD), bone percentage and trabecular thickness (Tb.Th) and increased alveolar bone porosity (p<0.05) and significantly reduced BALP serum levels, as well as radiographic density and Young's module of femur, compared to Naïve. There was a greater ABL in group GIOP+EP when compared to EP (p<0.05). GIOP+EP caused a greater decrease on ABMD, Tb.Th, bone percentage and increased bone porosity (p<0.05) and also presented a significant reduction in BALP levels (p<0.05), in radiographic density and in Young's module of femur compared to EP (p<0.05). CONCLUSIONS GIOP can potentiate the destructive effects of EP on alveolar bone and alter the systemic bone loss, by promoting bone resorption and reducing osteoblast activity.
Collapse
Affiliation(s)
- Luzia Hermínia Teixeira Sousa
- Post-graduation Program of Health Science, Medical School, Federal University of Ceará - Sobral, Avenida Comandante Maurocelio Rocha Pontes, 100 - Derby, Sobral, CE, 62.042-280, Brazil
| | - Eveline Valeriano Moura
- Post-graduation Program of Health Science, Medical School, Federal University of Ceará - Sobral, Avenida Comandante Maurocelio Rocha Pontes, 100 - Derby, Sobral, CE, 62.042-280, Brazil
| | - Ana Larissa Queiroz
- School of Dentistry, Federal University of Ceará - Sobral, R. Cel. Estanislau Frota - Centro, Sobral, CE, 62.010-560, Brazil
| | - Danielle Val
- Post Graduation Program RENORBIO, Federal University of Pernambuco, Av. Professor Morais Rego, 1235 - Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Hellíada Chaves
- Post-graduation Program of Health Science, Medical School, Federal University of Ceará - Sobral, Avenida Comandante Maurocelio Rocha Pontes, 100 - Derby, Sobral, CE, 62.042-280, Brazil; School of Dentistry, Federal University of Ceará - Sobral, R. Cel. Estanislau Frota - Centro, Sobral, CE, 62.010-560, Brazil
| | - Mario Lisboa
- Post-graduation Program of Morphological Science, Department of Morphology, Medical School, Federal University of Ceará - Fortaleza, Rua Delmiro de Farias, s/n - Rodolfo Teófilo, Fortaleza, CE, 60.430-170, Brazil
| | - Flávia Furlaneto
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto School of Dentistry, University of São Paulo, Av. do Café, s/n - Vila Amelia, Ribeirão Preto, São Paulo, 14050-904, Brazil, Brazil
| | - Gerly Anne Brito
- Post-graduation Program of Morphological Science, Department of Morphology, Medical School, Federal University of Ceará - Fortaleza, Rua Delmiro de Farias, s/n - Rodolfo Teófilo, Fortaleza, CE, 60.430-170, Brazil; Department of Morphology, Medical School, Federal University of Ceará - Fortaleza, Rua Delmiro de Farias, s/n - Rodolfo Teófilo, Fortaleza, CE, CEP 60.430-170, Brazil
| | - Paula Goes
- Post-graduation Program of Health Science, Medical School, Federal University of Ceará - Sobral, Avenida Comandante Maurocelio Rocha Pontes, 100 - Derby, Sobral, CE, 62.042-280, Brazil; Department of Pathology and Legal Medicine, Medical School, Federal University of Ceará - Fortaleza, Rua Monsenhor Furtado, S/N - Rodolfo Teófilo, Fortaleza, CE, 60.441-750, Brazil.
| |
Collapse
|
74
|
Ito M, Yamazaki S, Yamagami K, Kuno M, Morita Y, Okuma K, Nakamura K, Chida N, Inami M, Inoue T, Shirakami S, Higashi Y. A novel JAK inhibitor, peficitinib, demonstrates potent efficacy in a rat adjuvant-induced arthritis model. J Pharmacol Sci 2016; 133:25-33. [PMID: 28117214 DOI: 10.1016/j.jphs.2016.12.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/13/2016] [Accepted: 12/07/2016] [Indexed: 12/18/2022] Open
Abstract
The Janus kinase (JAK) family of tyrosine kinases is associated with various cytokine receptors. JAK1 and JAK3 play particularly important roles in the immune response, and their inhibition is expected to provide targeted immune modulation. Several oral JAK inhibitors have recently been developed for treating autoimmune diseases, including rheumatoid arthritis (RA). Here, we investigated the pharmacological effects of peficitinib (formerly known as ASP015K), a novel, chemically synthesized JAK inhibitor. We found that peficitinib inhibited JAK1 and JAK3 with 50% inhibitory concentrations of 3.9 and 0.7 nM, respectively. Peficitinib also inhibited IL-2-dependent T cell proliferation in vitro and STAT5 phosphorylation in vitro and ex vivo. Furthermore, peficitinib dose-dependently suppressed bone destruction and paw swelling in an adjuvant-induced arthritis model in rats via prophylactic or therapeutic oral dosing regimens. Peficitinib also showed efficacy in the model by continuous intraperitoneal infusion. Area under the concentration versus time curve (AUC) at 50% inhibition of paw swelling via intraperitoneal infusion was similar to exposure levels of AUC at 50% inhibition via oral administration, implying that AUC might be important for determining the therapeutic efficacy of peficitinib. These data suggest that peficitinib has therapeutic potential for the oral treatment of RA.
Collapse
Affiliation(s)
- Misato Ito
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan.
| | - Shunji Yamazaki
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Kaoru Yamagami
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Masako Kuno
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Yoshiaki Morita
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Kenji Okuma
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Koji Nakamura
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Noboru Chida
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Masamichi Inami
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Takayuki Inoue
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Shohei Shirakami
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Yasuyuki Higashi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| |
Collapse
|
75
|
Tong Y, Niu M, Du Y, Mei W, Cao W, Dou Y, Yu H, Du X, Yuan H, Zhao W. Aryl hydrocarbon receptor suppresses the osteogenesis of mesenchymal stem cells in collagen-induced arthritic mice through the inhibition of β-catenin. Exp Cell Res 2016; 350:349-357. [PMID: 28007558 DOI: 10.1016/j.yexcr.2016.12.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/17/2016] [Accepted: 12/18/2016] [Indexed: 12/13/2022]
Abstract
The contributions of aryl hydrocarbon receptor (Ahr) to the pathogenesis of rheumatoid arthritis (RA), particularly bone loss, have not been clearly explored. The imbalance between osteoblasts and osteoclasts is a major reason for bone loss. The dysfunction of osteoblasts, which are derived from mesenchymal stem cells (MSCs), induced bone erosion occurs earlier and is characterized as more insidious. Here, we showed that the nuclear expression and translocation of Ahr were both significantly increased in MSCs from collagen-induced arthritis (CIA) mice. The enhanced Ahr suppressed the mRNA levels of osteoblastic markers including Alkaline phosphatase (Alp) and Runt-related transcription factor 2 (Runx2) in the differentiation of MSCs to osteoblasts in CIA. The 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated activation of Ahr dose-dependently suppressed the expression of osteoblastic markers. In addition, the expression of β-catenin was reduced in CIA MSCs compared with control, and the TCDD-mediated activation of the Ahr significantly inhibited β-catenin expression. The Wnt3a-induced the activation of Wnt/β-catenin pathway partly rescued the osteogenesis decline induced by TCDD. Taken together, these results indicate that activated Ahr plays a negative role in CIA MSCs osteogenesis, possibly by suppressing the expression of β-catenin.
Collapse
Affiliation(s)
- Yulong Tong
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China
| | - Menglin Niu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China; Department of Blood Transfusion, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing 100142, PR China
| | - Yuxuan Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China
| | - Wentong Mei
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China
| | - Wei Cao
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China
| | - Yunpeng Dou
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China
| | - Haitao Yu
- Department of Clinical Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu Province 730000, PR China
| | - Xiaonan Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China
| | - Huihui Yuan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China.
| | - Wenming Zhao
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing 100069, PR China.
| |
Collapse
|
76
|
Fu L, Pan F, Jiao Y. Crocin inhibits RANKL-induced osteoclast formation and bone resorption by suppressing NF-κB signaling pathway activation. Immunobiology 2016; 222:597-603. [PMID: 27871781 DOI: 10.1016/j.imbio.2016.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 01/11/2023]
Abstract
Crocin is a dietary compound with antioxidant and anti-inflammatory properties, but its effects on bone resorption have not been well characterized. Here we address this issue by examining the direct effects of crocin on osteoclast cells in vitro. Osteoclastogenesis was induced by RANKL (receptor activator of NF-κB ligand) in mouse bone marrow-derived macrophages in the absence or presence of crocin at various concentrations. Further, the bone resorption activity of mature osteoclast treated with crocin was assessed by pit assay. Without altering cell viability, crocin was shown to inhibit the differentiation and function of osteoclast cells in a dose-dependent manner. Mechanistically, RANKL-induced NF-κB and NFATc1 activation, the critical signaling pathways for osteoclast differentiation and function, were both repressed by crocin in bone marrow-derived macrophages. Thus, crocin suppresses osteoclastogenesis through direct inhibition of intracellular molecular pathways, which may contribute to future development of anti-bone resorption treatment.
Collapse
Affiliation(s)
- Lijia Fu
- Department of Preparation Room, Daqing Oilfield General Hospital, Daqing 163001, Heilongjiang Province, China
| | - Fang Pan
- Department of Rheumatology, Daqing Oilfield General Hospital, Daqing 163001, Heilongjiang Province, China
| | - Yong Jiao
- Department of Orthopaedics, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, No. 5 Haiyun Cang, Dongzhimen District, Beijing 100070, China.
| |
Collapse
|
77
|
Szeremeta A, Jura-Półtorak A, Komosińska-Vassev K, Zoń-Giebel A, Kapołka D, Olczyk K. The association between insulin-like growth factor 1 (IGF-1), IGF-binding proteins (IGFBPs), and the carboxyterminal propeptide of type I procollagen (PICP) in pre- and postmenopausal women with rheumatoid arthritis. Scand J Rheumatol 2016; 46:171-179. [PMID: 27775453 DOI: 10.1080/03009742.2016.1203020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVES To assess the association between plasma levels of the insulin-like growth factor (IGF) system including IGF-1, IGF-binding proteins (IGFBPs) including IGFBP-1, total (t-)IGFBP-3 and functional (f-)IGFBP-3, and the carboxyterminal propeptide of type I procollagen (PICP) in pre- and postmenopausal women with rheumatoid arthritis (RA). METHOD Plasma concentrations of IGF-1, IGFBP-1, t-IGFBP-3, f-IGFBP-3, and PICP were measured by immunoassay. RESULTS No significant difference was observed in plasma IGF-1 levels between pre- and postmenopausal subjects. Plasma levels of IGFBP-1 were elevated in RA. PICP and f-IGFBP-3 were greatly affected by menopausal status. Of the three IGFBPs tested, only f-IGFBP-3 plasma levels in RA women correlated negatively with age and disease duration. A positive correlation was demonstrated between PICP and erythrocyte sedimentation rate (ESR) in RA. Moreover, there was no correlation between PICP and IGF-1 and any of the IGFBPs in RA women. CONCLUSIONS Considerable disruption of the IGF system in RA was found to be related to disease activity and duration. Changes in the IGF-IGFBP axis and PICP levels were different in pre- and postmenopausal women with RA. Elevated plasma PICP concentrations may indicate an increased rate of bone formation in postmenopausal RA women. Additionally, the observed changes in the IGF/IGFBP system did not affect bone formation during RA.
Collapse
Affiliation(s)
- A Szeremeta
- a Department of Clinical Chemistry and Laboratory Diagnostics , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice , Sosnowiec , Poland
| | - A Jura-Półtorak
- a Department of Clinical Chemistry and Laboratory Diagnostics , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice , Sosnowiec , Poland
| | - K Komosińska-Vassev
- a Department of Clinical Chemistry and Laboratory Diagnostics , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice , Sosnowiec , Poland
| | - A Zoń-Giebel
- b Department of Rheumatology , Silesian Centre for Rheumatology, Rehabilitation, and Prevention of Disability of Gen. Jerzy Ziętek in Ustroń , Ustroń , Poland
| | - D Kapołka
- b Department of Rheumatology , Silesian Centre for Rheumatology, Rehabilitation, and Prevention of Disability of Gen. Jerzy Ziętek in Ustroń , Ustroń , Poland
| | - K Olczyk
- a Department of Clinical Chemistry and Laboratory Diagnostics , School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice , Sosnowiec , Poland
| |
Collapse
|
78
|
Hu X, Garcia M, Weng L, Jung X, Murakami JL, Kumar B, Warden CD, Todorov I, Chen CC. Identification of a common mesenchymal stromal progenitor for the adult haematopoietic niche. Nat Commun 2016; 7:13095. [PMID: 27721421 PMCID: PMC5062560 DOI: 10.1038/ncomms13095] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/01/2016] [Indexed: 12/13/2022] Open
Abstract
Microenvironment cues received by haematopoietic stem cells (HSC) are important in regulating the choice between self-renewal and differentiation. On the basis of the differential expression of cell-surface markers, here we identify a mesenchymal stromal progenitor hierarchy, where CD45−Ter119−CD31−CD166−CD146−Sca1+(Sca1+) progenitors give rise to CD45−Ter119−CD31−CD166−CD146+(CD146+) intermediate and CD45−Ter119−CD31−CD166+CD146−(CD166+) mature osteo-progenitors. All three progenitors preserve HSC long-term multi-lineage reconstitution capability in vitro; however, their in vivo fates are different. Post-transplantation, CD146+ and CD166+ progenitors form bone only. While Sca1+ progenitors produce CD146+, CD166+ progenitors, osteocytes and CXCL12-producing stromal cells. Only Sca1+ progenitors are capable of homing back to the marrow post-intravenous infusion. Ablation of Sca1+ progenitors results in a decrease of all three progenitor populations as well as haematopoietic stem/progenitor cells. Moreover, suppressing production of KIT-ligand in Sca1+ progenitors inhibits their ability to support HSCs. Our results indicate that Sca1+ progenitors, through the generation of both osteogenic and stromal cells, provide a supportive environment for hematopoiesis. How the environment of the niche regulates haematopoietic stem cells (HSC) is unclear. Here, the authors identify a mesenchymal stromal progenitor hierarchy and identify Sca1+ cells as common progenitors for mesenchymal stromal cells in the adult niche that provide a supportive environment for hematopoiesis.
Collapse
Affiliation(s)
- Xingbin Hu
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA.,Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 7100032, PR China
| | - Mayra Garcia
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Lihong Weng
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Xiaoman Jung
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Jodi L Murakami
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA.,Irell &Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010, USA
| | - Bijender Kumar
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Charles D Warden
- Bioinformatics Core, Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Ivan Todorov
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA
| | - Ching-Cheng Chen
- Divison of Hematopoietic Stem Cell and Leukemia Research, Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Duarte, California 91010, USA.,Irell &Manella Graduate School of Biological Sciences, City of Hope, Duarte, California 91010, USA
| |
Collapse
|
79
|
Bugatti S, Bogliolo L, Vitolo B, Manzo A, Montecucco C, Caporali R. Anti-citrullinated protein antibodies and high levels of rheumatoid factor are associated with systemic bone loss in patients with early untreated rheumatoid arthritis. Arthritis Res Ther 2016; 18:226. [PMID: 27716332 PMCID: PMC5052789 DOI: 10.1186/s13075-016-1116-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/13/2016] [Indexed: 01/19/2023] Open
Abstract
Background Autoantibodies such as anti-citrullinated protein antibodies (ACPA) are major risk factors for articular bone destruction from the earliest phases of rheumatoid arthritis (RA). The aim of the current study was to determine whether RA-associated autoantibodies also impact on systemic bone loss in patients with early disease. Methods Systemic bone mineral density (BMD) was measured in the lumbar spine and the hip in 155 consecutive treatment-naïve patients with early RA (median symptom duration 13 weeks). Demographic and disease-specific parameters, including clinical disease activity, ultrasonographic (US) examination of the hands and wrists, radiographic scoring of joint damage, ACPA and rheumatoid factor (RF) levels were recorded from all patients. Reduced BMD was defined as Z score ≤ -1 SD and analysed in relation to disease-related characteristics and autoantibody subgroups. Results Reduced BMD was found in 25.5 % of the patients in the spine and 19.4 % in the hip. Symptom duration, clinical and US disease activity, functional disability and radiographic damage did not significantly impact on spine and hip BMD loss in regression analyses adjusted for possible confounders (age, gender, menopausal status, current smoking, body mass index). In contrast, ACPA positivity (at any level) negatively affected the spine Z-score (adjusted OR (95 % CI) 2.76 (1.19 to 6.42)); the hip Z score was affected by high titres only (adjusted OR (95 % CI) 2.96 (1.15 to 7.66)). The association of ACPA with reduced BMD in the spine was confirmed even at low levels of RF (adjusted OR (95 % CI) 2.65 (1.01 to 7.24)), but was further increased by concomitant high RF (adjusted OR (95 % CI) 3.38 (1.11 to 10.34)). In contrast, Z scores in the hip were significantly reduced only in association with high ACPA and RF (adjusted OR (95 % CI) 4.96 (1.48 to 16.64)). Conclusions Systemic BMD in patients with early RA is reduced in relation with ACPA positivity and high RF levels. This finding supports the notion that RA-associated autoimmunity may have a direct causative role in bone remodeling.
Collapse
Affiliation(s)
- Serena Bugatti
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Early Arthritis Clinic, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Piazzale Golgi 19, 27100, Pavia, Italy
| | - Laura Bogliolo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Early Arthritis Clinic, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Piazzale Golgi 19, 27100, Pavia, Italy
| | - Barbara Vitolo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Early Arthritis Clinic, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Piazzale Golgi 19, 27100, Pavia, Italy
| | - Antonio Manzo
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Early Arthritis Clinic, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Piazzale Golgi 19, 27100, Pavia, Italy
| | - Carlomaurizio Montecucco
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Early Arthritis Clinic, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Piazzale Golgi 19, 27100, Pavia, Italy.
| | - Roberto Caporali
- Rheumatology and Translational Immunology Research Laboratories (LaRIT) and Early Arthritis Clinic, Division of Rheumatology, IRCCS Policlinico San Matteo Foundation/University of Pavia, Piazzale Golgi 19, 27100, Pavia, Italy
| |
Collapse
|
80
|
Alves CH, Farrell E, Vis M, Colin EM, Lubberts E. Animal Models of Bone Loss in Inflammatory Arthritis: from Cytokines in the Bench to Novel Treatments for Bone Loss in the Bedside-a Comprehensive Review. Clin Rev Allergy Immunol 2016; 51:27-47. [PMID: 26634933 PMCID: PMC4961736 DOI: 10.1007/s12016-015-8522-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Throughout life, bone is continuously remodelled. Bone is formed by osteoblasts, from mesenchymal origin, while osteoclasts induce bone resorption. This process is tightly regulated. During inflammation, several growth factors and cytokines are increased inducing osteoclast differentiation and activation, and chronic inflammation is a condition that initiates systemic bone loss. Rheumatoid arthritis (RA) is a chronic inflammatory auto-immune disease that is characterised by active synovitis and is associated with early peri-articular bone loss. Peri-articular bone loss precedes focal bone erosions, which may progress to bone destruction and disability. The incidence of generalised osteoporosis is associated with the severity of arthritis in RA and increased osteoporotic vertebral and hip fracture risk. In this review, we will give an overview of different animal models of inflammatory arthritis related to RA with focus on bone erosion and involvement of pro-inflammatory cytokines. In addition, a humanised endochondral ossification model will be discussed, which can be used in a translational approach to answer osteoimmunological questions.
Collapse
Affiliation(s)
- C Henrique Alves
- Department of Rheumatology, Erasmus MC, University Medical Center, Wytemaweg 80, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center, Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Marijn Vis
- Department of Rheumatology, Erasmus MC, University Medical Center, Wytemaweg 80, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Edgar M Colin
- Department of Rheumatology, Erasmus MC, University Medical Center, Wytemaweg 80, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Rheumatology, ZGT Almelo, Zilvermeeuw 1, 7600 SZ, Almelo, The Netherlands
| | - Erik Lubberts
- Department of Rheumatology, Erasmus MC, University Medical Center, Wytemaweg 80, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
81
|
Xie Z, Li J, Wang P, Li Y, Wu X, Wang S, Su H, Deng W, Liu Z, Cen S, Ouyang Y, Wu Y, Shen H. Differential Expression Profiles of Long Noncoding RNA and mRNA of Osteogenically Differentiated Mesenchymal Stem Cells in Ankylosing Spondylitis. J Rheumatol 2016; 43:1523-31. [PMID: 27182066 DOI: 10.3899/jrheum.151181] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVE We previously demonstrated that mesenchymal stem cells (MSC) from patients with ankylosing spondylitis (AS; ASMSC) have a greater osteogenic differentiation capacity than MSC from healthy donors (HDMSC) and that this difference underlies the pathogenesis of pathological osteogenesis in AS. Here we compared expression levels of long noncoding RNA (lncRNA) and mRNA between osteogenically differentiated ASMSC and HDMSC and explored the precise mechanism underlying abnormal osteogenic differentiation in ASMSC. METHODS HDMSC and ASMSC were induced with osteogenic differentiation medium for 10 days. Microarray analyses were then performed to identify lncRNA and mRNA differentially expressed between HDMSC and ASMSC, which were then subjected to bioinformatics analysis and confirmed by quantitative real-time PCR (qRT-PCR) assays. In addition, coding-non-coding gene co-expression (CNC) networks were constructed to examine the relationships between the lncRNA and mRNA expression patterns. RESULTS A total of 520 lncRNA and 665 mRNA were differentially expressed in osteogenically differentiated ASMSC compared with HDMSC. Bioinformatics analysis revealed 64 signaling pathways with significant differences, including transforming growth factor-β signaling. qRT-PCR assays confirmed the reliability of the microarray data. The CNC network indicated that 4 differentially expressed lncRNA, including lnc-ZNF354A-1, lnc-LIN54-1, lnc-FRG2C-3, and lnc-USP50-2 may be involved in the abnormal osteogenic differentiation of ASMSC. CONCLUSION Our study characterized the differential lncRNA and mRNA expression profiles of osteogenically differentiated ASMSC and identified 4 lncRNA that may participate in the abnormal osteogenic differentiation of ASMSC. These results provide insight into the pathogenesis of pathological osteogenesis in AS.
Collapse
Affiliation(s)
- Zhongyu Xie
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Jinteng Li
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Peng Wang
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Yuxi Li
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Xiaohua Wu
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Shan Wang
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Hongjun Su
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Wen Deng
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Zhenhua Liu
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Shuizhong Cen
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Yi Ouyang
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Yanfeng Wu
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Huiyong Shen
- From the Department of Orthopedics, and the Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P.R. China.Z. Xie, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; J. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; P. Wang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Li, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; X. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Wang, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Su, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; W. Deng, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Z. Liu, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; S. Cen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Ouyang, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; Y. Wu, MD, Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University; H. Shen, MD, Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University.
| |
Collapse
|
82
|
Wang N, Zhou Z, Wu T, Liu W, Yin P, Pan C, Yu X. TNF-α-induced NF-κB activation upregulates microRNA-150-3p and inhibits osteogenesis of mesenchymal stem cells by targeting β-catenin. Open Biol 2016; 6:150258. [PMID: 26935950 PMCID: PMC4821240 DOI: 10.1098/rsob.150258] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/05/2016] [Indexed: 12/30/2022] Open
Abstract
Although systemic or local inflammation, commonly featured by cytokine activation, is implicated in patients with bone loss, the underlying mechanisms are still elusive. As microRNAs (miR), a class of small non-coding RNAs involved in essential physiological processes, have been found in bone cells, we aimed to investigate the role of miR for modulating osteogenesis in inflammatory milieu using human bone marrow mesenchymal stem cells (hBM-MSCs). Induced by proinflammatory cytokine TNF-α, miR-150-3p was identified as a key player in suppressing osteogenic differentiation through downregulating β-catenin, a transcriptional co-activator promoting bone formation. TNF-α treatment increased the levels of miR-150-3p, which directly targeted the 3'-UTR of β-catenin mRNA and in turn repressed its expression. In addition, we observed that miR-150-3p expression was increased by TNF-α via IKK-dependent NF-κB signalling. There are three putative NF-κB binding sites in the promoter region of miR-150, and we identified -686 region as the major NF-κB binding site for stimulation of miR-150 expression by TNF-α. Finally, the osteogenic differentiation of hBM-MSCs was inhibited by either miR-150-3p overexpression or TNF-α treatment, which was prevented by anti-miR-150-3p oligonucleotides. Taken together, our data suggested that miR-150-3p integrated inflammation signalling and osteogenic differentiation and may contribute to the inhibition effects of inflammation on bone formation, thus expanding the pathophysiological functions of microRNAs in bone diseases.
Collapse
Affiliation(s)
- Nan Wang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, Shanghai, People's Republic of China Department of Emergency, the first affiliated hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zubin Zhou
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Tianyi Wu
- Department of Orthopaedics and Traumatology, Lui Che Woo Institute of Innovative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China
| | - Wei Liu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Peipei Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Chenhao Pan
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Xiaowei Yu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| |
Collapse
|
83
|
Su CM, Huang CY, Tang CH. Characteristics of resistin in rheumatoid arthritis angiogenesis. Biomark Med 2016; 10:651-60. [PMID: 26867862 DOI: 10.2217/bmm.15.125] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipokines have been reported to be involved in the regulation of various physiological processes, including the immune response. Rheumatoid arthritis (RA) is an example of a systemic immune disease that causes chronic inflammation of the synovium and bone destruction in the joint. Recent therapeutic strategies based on the understanding of the role of cytokines and cellular mechanisms in RA have improved our understanding of angiogenesis. On the other hand, endogenous endothelial progenitor cells, which are a population isolated from peripheral blood monocytes have recently been identified as a homing target for pro-angiogeneic factor and vessel formation. In this review, we summarize the effects of common adipokines, such as adiponectin, leptin and resistin in RA pathogenesis and discuss other potential mechanisms of relevance for the therapeutic treatment of RA.
Collapse
Affiliation(s)
- Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
84
|
Luo Y. A biomechanical sorting of clinical risk factors affecting osteoporotic hip fracture. Osteoporos Int 2016; 27:423-39. [PMID: 26361947 DOI: 10.1007/s00198-015-3316-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 09/03/2015] [Indexed: 02/07/2023]
Abstract
Osteoporotic fracture has been found associated with many clinical risk factors, and the associations have been explored dominantly by evidence-based and case-control approaches. The major challenges emerging from the studies are the large number of the risk factors, the difficulty in quantification, the incomplete list, and the interdependence of the risk factors. A biomechanical sorting of the risk factors may shed lights on resolving the above issues. Based on the definition of load-strength ratio (LSR), we first identified the four biomechanical variables determining fracture risk, i.e., the risk of fall, impact force, bone quality, and bone geometry. Then, we explored the links between the FRAX clinical risk factors and the biomechanical variables by looking for evidences in the literature. To accurately assess fracture risk, none of the four biomechanical variables can be ignored and their values must be subject-specific. A clinical risk factor contributes to osteoporotic fracture by affecting one or more of the biomechanical variables. A biomechanical variable represents the integral effect from all the clinical risk factors linked to the variable. The clinical risk factors in FRAX mostly stand for bone quality. The other three biomechanical variables are not adequately represented by the clinical risk factors. From the biomechanical viewpoint, most clinical risk factors are interdependent to each other as they affect the same biomechanical variable(s). As biomechanical variables must be expressed in numbers before their use in calculating LSR, the numerical value of a biomechanical variable can be used as a gauge of the linked clinical risk factors to measure their integral effect on fracture risk, which may be more efficient than to study each individual risk factor.
Collapse
Affiliation(s)
- Y Luo
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB, Canada.
- Department of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada.
- Department of Anatomy, South Medical University, Guangzhou, China.
| |
Collapse
|
85
|
Liu W, Fan JB, Xu DW, Zhang J, Cui ZM. Epigallocatechin-3-gallate protects against tumor necrosis factor alpha induced inhibition of osteogenesis of mesenchymal stem cells. Exp Biol Med (Maywood) 2016; 241:658-66. [PMID: 26748399 DOI: 10.1177/1535370215624020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/26/2015] [Indexed: 11/16/2022] Open
Abstract
Anabolic bone accruement through osteogenic differentiation is important for the maintenance of physiological bone mass and often disrupted in various inflammatory diseases. Epigallocatechin-3-gallate, as an antioxidant and anti-inflammatory agent, has been suggested for potential therapeutic use in this context, possibly by the inhibition of bone resorption as well as the enhancement of bone formation through directly activating osteoblast differentiation. However, the reported effects of epigallocatechin-3-gallate modulating osteoblast differentiation are mixed, and the underlying molecular mechanism is still elusive. Moreover, there is limited information regarding the effects of epigallocatechin-3-gallate on osteogenic potential of mesenchymal stem cell in inflammation. Here, we examined the in vitro osteogenic differentiation of human mesenchymal stem cells. We found that the cell viability and osteoblast differentiation of human bone marrow-derived mesenchymal stem cells are significantly inhibited by inflammatory cytokine TNFα treatment. Epigallocatechin-3-gallate is able to enhance the cell viability and osteoblast differentiation of mesenchymal stem cells and is capable of reversing the TNFα-induced inhibition. Notably, only low doses of epigallocatechin-3-gallate have such benefits, which potentially act through the inhibition of NF-κB signaling that is stimulated by TNFα. These data altogether clarify the controversy on epigallocatechin-3-gallate promoting osteoblast differentiation and further provide molecular basis for the putative clinical use of epigallocatechin-3-gallate in stem cell-based bone regeneration for inflammatory bone loss diseases, such as rheumatoid arthritis and prosthetic osteolysis.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jian-Bo Fan
- Department of Orthopedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Da-Wei Xu
- Department of Orthopedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jie Zhang
- School of Medicine, Nantong University, Nantong 226019, China The first two authors contributed equally to this work
| | - Zhi-Ming Cui
- Department of Orthopedics, The Second Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
86
|
Okragly AJ, Hamang MJ, Pena EA, Baker HE, Bullock HA, Lucchesi J, Martin AP, Ma YL, Benschop RJ. Elevated levels of Interleukin (IL)-33 induce bone pathology but absence of IL-33 does not negatively impact normal bone homeostasis. Cytokine 2016; 79:66-73. [PMID: 26771472 DOI: 10.1016/j.cyto.2015.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/05/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
Interleukin (IL)-33 is a member of the IL-1 family. IL-33 effects are mediated through its receptor, ST2 and IL-1RAcP, and its signaling induces the production of a number of pro-inflammatory mediators, including TNFα, IL-1β, IL-6, and IFN-γ. There are conflicting reports on the role of IL-33 in bone homeostasis, with some demonstrating a bone protective role for IL-33 whilst others show that IL-33 induces inflammatory arthritis with concurrent bone destruction. To better clarify the role IL-33 plays in bone biology in vivo, we studied IL-33 KO mice as well as mice in which the cytokine form of IL-33 was overexpressed. Mid-femur cortical bone mineral density (BMD) and bone strength were similar in the IL-33 KO mice compared to WT animals during the first 8months of life. However, in the absence of IL-33, we observed higher BMD in lumbar vertebrae and distal femur in female mice. In contrast, overexpression of IL-33 resulted in a marked and rapid reduction of bone volume, mineral density and strength. Moreover, this was associated with a robust increase in inflammatory cytokines (including IL-6 and IFN-γ), suggesting the bone pathology could be a direct effect of IL-33 or an indirect effect due to the induction of other mediators. Furthermore, the detrimental bone effects were accompanied by increases in osteoclast number and the bone resorption marker of C-terminal telopeptide collagen-I (CTX-I). Together, these results demonstrate that absence of IL-33 has no negative consequences in normal bone homeostasis while high levels of circulating IL-33 contributes to pathological bone loss.
Collapse
Affiliation(s)
- Angela J Okragly
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Matthew J Hamang
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Emily A Pena
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Hana E Baker
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Heather A Bullock
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jonathan Lucchesi
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Andrea P Martin
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Y Linda Ma
- Musculoskeletal-Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Robert J Benschop
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA.
| |
Collapse
|
87
|
Su CM, Lee WL, Hsu CJ, Lu TT, Wang LH, Xu GH, Tang CH. Adiponectin Induces Oncostatin M Expression in Osteoblasts through the PI3K/Akt Signaling Pathway. Int J Mol Sci 2015; 17:ijms17010029. [PMID: 26712749 PMCID: PMC4730275 DOI: 10.3390/ijms17010029] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/16/2015] [Accepted: 12/22/2015] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA), a common autoimmune disorder, is associated with a chronic inflammatory response and unbalanced bone metabolism within the articular microenvironment. Adiponectin, an adipokine secreted by adipocytes, is involved in multiple functions, including lipid metabolism and pro-inflammatory activity. However, the mechanism of adiponectin performance within arthritic inflammation remains unclear. In this study, we observed the effect of adiponectin on the expression of oncostatin M (OSM), a pro-inflammatory cytokine, in human osteoblastic cells. Pretreatment of cells with inhibitors of phosphatidylinositol 3-kinase (PI3K), Akt, and nuclear factor (NF)-κB reduced the adiponectin-induced OSM expression in osteoblasts. Stimulation of the cells with adiponectin increased phosphorylation of PI3K, Akt, and p65. Adiponectin treatment of osteoblasts increased OSM-luciferase activity and p65 binding to NF-κB on the OSM promoter. Our results indicate that adiponectin increased OSM expression via the PI3K, Akt, and NF-κB signaling pathways in osteoblastic cells, suggesting that adiponectin is a novel target for arthritis treatment.
Collapse
Affiliation(s)
- Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang 322100, China.
| | - Wei-Lin Lee
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| | - Chin-Jung Hsu
- School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan.
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Ting-Ting Lu
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang 322100, China.
| | - Li-Hong Wang
- Department of Orthopedics, Dongyang Peoples' Hospital, Dongyang 322100, China.
| | - Guo-Hong Xu
- Department of Orthopedics, Dongyang Peoples' Hospital, Dongyang 322100, China.
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan.
- Department of Biotechnology, College of Health Science, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
88
|
Xu X, Zheng L, Bian Q, Xie L, Liu W, Zhen G, Crane JL, Zhou X, Cao X. Aberrant Activation of TGF-β in Subchondral Bone at the Onset of Rheumatoid Arthritis Joint Destruction. J Bone Miner Res 2015; 30:2033-43. [PMID: 25967237 PMCID: PMC4809636 DOI: 10.1002/jbmr.2550] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/23/2015] [Accepted: 05/07/2015] [Indexed: 02/05/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that often leads to joint destruction. A myriad of drugs targeting the immune abnormalities and downstream inflammatory cascades have been developed, but the joint destruction is not effectively halted. Here we report that aberrant activation of TGF-β in the subchondral bone marrow by immune response increases osteoprogenitors and uncoupled bone resorption and formation in RA mouse/rat models. Importantly, either systemic or local blockade of TGF-β activity in the subchondral bone attenuated articular cartilage degeneration in RA. Moreover, conditional deletion of TGF-β receptor II (Tgfbr2) in nestin-positive cells also effectively halted progression of RA joint destruction. Our data demonstrate that aberrant activation of TGF-β in the subchondral bone is involved at the onset of RA joint cartilage degeneration. Thus, modulation of subchondral bone TGF-β activity could be a potential therapy for RA joint destruction.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qin Bian
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute of Spine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenlong Liu
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China
| | - Gehua Zhen
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janet L Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
89
|
Tatman PD, Gerull W, Sweeney-Easter S, Davis JI, Gee AO, Kim DH. Multiscale Biofabrication of Articular Cartilage: Bioinspired and Biomimetic Approaches. TISSUE ENGINEERING PART B-REVIEWS 2015. [PMID: 26200439 DOI: 10.1089/ten.teb.2015.0142] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Articular cartilage is the load-bearing tissue found inside all articulating joints of the body. It vastly reduces friction and allows for smooth gliding between contacting surfaces. The structure of articular cartilage matrix and cellular composition is zonal and is important for its mechanical properties. When cartilage becomes injured through trauma or disease, it has poor intrinsic healing capabilities. The spectrum of cartilage injury ranges from isolated areas of the joint to diffuse breakdown and the clinical appearance of osteoarthritis. Current clinical treatment options remain limited in their ability to restore cartilage to its normal functional state. This review focuses on the evolution of biomaterial scaffolds that have been used for functional cartilage tissue engineering. In particular, we highlight recent developments in multiscale biofabrication approaches attempting to recapitulate the complex 3D matrix of native articular cartilage tissue. Additionally, we focus on the application of these methods to engineering each zone of cartilage and engineering full-thickness osteochondral tissues for improved clinical implantation. These methods have shown the potential to control individual cell-to-scaffold interactions and drive progenitor cell differentiation into a chondrocyte lineage. The use of these bioinspired nanoengineered scaffolds hold promise for recreation of structure and function on the whole tissue level and may represent exciting new developments for future clinical applications for cartilage injury and restoration.
Collapse
Affiliation(s)
- Philip David Tatman
- 1 Department of Bioengineering, University of Washington , Seattle, Washington
| | - William Gerull
- 1 Department of Bioengineering, University of Washington , Seattle, Washington
| | - Sean Sweeney-Easter
- 1 Department of Bioengineering, University of Washington , Seattle, Washington
| | - Jeffrey Isaac Davis
- 1 Department of Bioengineering, University of Washington , Seattle, Washington
| | - Albert O Gee
- 2 Department of Orthopedics and Sports Medicine, University of Washington , Seattle, Washington
| | - Deok-Ho Kim
- 1 Department of Bioengineering, University of Washington , Seattle, Washington.,3 Institute for Stem Cell and Regenerative Medicine, University of Washington , Seattle, Washington
| |
Collapse
|
90
|
Jang WD, Kim JT, Son HY, Park SY, Cho YS, Koo TS, Lee H, Kang NS. Discovery of Tyk2 inhibitors via the virtual site-directed fragment-based drug design. Bioorg Med Chem Lett 2015; 25:3947-52. [DOI: 10.1016/j.bmcl.2015.07.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/29/2015] [Accepted: 07/15/2015] [Indexed: 11/30/2022]
|
91
|
Su CM, Chiang YC, Huang CY, Hsu CJ, Fong YC, Tang CH. Osteopontin Promotes Oncostatin M Production in Human Osteoblasts: Implication of Rheumatoid Arthritis Therapy. THE JOURNAL OF IMMUNOLOGY 2015; 195:3355-64. [PMID: 26304992 DOI: 10.4049/jimmunol.1403191] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 07/23/2015] [Indexed: 12/21/2022]
Abstract
Accumulating evidence indicates that subchondral bone might play an essential role in rheumatoid arthritis (RA). Osteopontin (OPN) induces the production of an important proinflammatory cytokine involved in the pathogenesis of RA. This study evaluated the activation of oncostatin M (OSM) by OPN in human primary osteoblasts to understand RA pathogenesis and characterized the intracellular signaling pathways involved in this activation. Quantitative PCR, ELISA, and Western blot results indicated that stimulation of human primary osteoblasts with OPN induces OSM expression through αvβ3 integrin/c-Src/platelet-derived growth factor receptor transactivation/MEK/ERK. Treatment of osteoblasts with OPN also increased c-Jun phosphorylation, AP-1 luciferase activity, and c-Jun binding to the AP-1 element on the OSM promoter, as demonstrated using chromatin immunoprecipitation assay. Moreover, inhibition of OPN expression using lentiviral-OPN short hairpin RNA resulted in the amelioration of articular swelling, cartilage erosion, and OSM expression in the ankle joint of mice with collagen-induced arthritis as shown using microcomputed tomography and immunohistochemistry staining. Our results imply that OSM expression in osteoblasts increases in response to OPN-induced inflammation in vitro. Finally, lentiviral-OPN short hairpin RNA ameliorates the inflammatory response and bone destruction in mice with collagen-induced arthritis. Therefore, OPN may be a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China 322100; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 40466
| | - Yi-Chun Chiang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 40466
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan 40466; Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan 65142
| | - Chin-Jung Hsu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan 40466; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan 40466
| | - Yi-Chin Fong
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan 40466; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan 40466
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 40466; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan 40466; and Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan 40466
| |
Collapse
|
92
|
Orosa B, Martínez P, González A, Guede D, Caeiro JR, Gómez-Reino JJ, Conde C. Effect of lysophosphatidic acid receptor inhibition on bone changes in ovariectomized mice. J Bone Miner Metab 2015; 33:383-91. [PMID: 24994065 DOI: 10.1007/s00774-014-0607-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/20/2014] [Indexed: 01/23/2023]
Abstract
Pharmacological inhibition of signaling through lysophosphatidic acid (LPA) receptors reduces bone erosions in an experimental model of arthritis by mechanisms involving reduced osteoclast differentiation and bone resorption and increased differentiation of osteoblasts and bone mineralization. These results led us to hypothesize that LPA receptor inhibition would be beneficial in osteoporosis. Our aim was to test this hypothesis with the LPA receptor antagonist, Ki16425, in ovariectomized mice, a model of postmenopausal osteoporosis. Ovariectomized mice treated with Ki16425 showed bone loss similar to that observed in the controls. Osteoblast markers, Alpl, Bglap and Col1a1, were increased at the mRNA level but no changes were detected in serum. No additional difference was observed in the Ki16425-treated mice relative to the ovariectomized controls with regard to osteoclast function markers or assays of matrix mineralization or osteoclast differentiation. Thus, pharmacological inhibition of LPA receptor was not beneficial for preventing bone loss in ovariectomized mice, indicating that its favorable effect on bone remodeling is less general than hypothesized.
Collapse
Affiliation(s)
- Beatriz Orosa
- Laboratorio de Investigación 8 y Servicio de Reumatología, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), SERGAS, Travesía da Choupana s/n, Santiago de Compostela, 15706, Spain
| | | | | | | | | | | | | |
Collapse
|
93
|
Kassem A, Henning P, Lundberg P, Souza PPC, Lindholm C, Lerner UH. Porphyromonas gingivalis Stimulates Bone Resorption by Enhancing RANKL (Receptor Activator of NF-κB Ligand) through Activation of Toll-like Receptor 2 in Osteoblasts. J Biol Chem 2015; 290:20147-58. [PMID: 26085099 DOI: 10.1074/jbc.m115.655787] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Indexed: 12/18/2022] Open
Abstract
Periodontitis has been associated with rheumatoid arthritis. In experimental arthritis, concomitant periodontitis caused by oral infection with Porphyromonas gingivalis enhances articular bone loss. The aim of this study was to investigate how lipopolysaccharide (LPS) from P. gingivalis stimulates bone resorption. The effects by LPS P. gingivalis and four other TLR2 ligands on bone resorption, osteoclast formation, and gene expression in wild type and Tlr2-deficient mice were assessed in ex vivo cultures of mouse parietal bones and in an in vivo model in which TLR2 agonists were injected subcutaneously over the skull bones. LPS P. gingivalis stimulated mineral release and matrix degradation in the parietal bone organ cultures by increasing differentiation and formation of mature osteoclasts, a response dependent on increased RANKL (receptor activator of NF-κB ligand). LPS P. gingivalis stimulated RANKL in parietal osteoblasts dependent on the presence of TLR2 and through a MyD88 and NF-κB-mediated mechanism. Similarly, the TLR2 agonists HKLM, FSL1, Pam2, and Pam3 stimulated RANKL in osteoblasts and parietal bone resorption. LPS P. gingivalis and Pam2 robustly enhanced osteoclast formation in periosteal/endosteal cell cultures by increasing RANKL. LPS P. gingivalis and Pam2 also up-regulated RANKL and osteoclastic genes in vivo, resulting in an increased number of periosteal osteoclasts and immense bone loss in wild type mice but not in Tlr2-deficient mice. These data demonstrate that LPS P. gingivalis stimulates periosteal osteoclast formation and bone resorption by stimulating RANKL in osteoblasts via TLR2. This effect might be important for periodontal bone loss and for the enhanced bone loss seen in rheumatoid arthritis patients with concomitant periodontal disease.
Collapse
Affiliation(s)
- Ali Kassem
- From the Department of Molecular Periodontology, Umeå University, 90187 Umeå, Sweden
| | - Petra Henning
- the Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition at Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Pernilla Lundberg
- From the Department of Molecular Periodontology, Umeå University, 90187 Umeå, Sweden
| | - Pedro P C Souza
- From the Department of Molecular Periodontology, Umeå University, 90187 Umeå, Sweden, the Department of Physiology and Pathology, Araraquara School of Dentistry, University Estudual Paulista (UNESP), Araraquara, Brazil 14801-903, and
| | - Catharina Lindholm
- the Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition at Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, 413 45 Gothenburg, Sweden, the Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, 403 50 Gothenburg, Sweden
| | - Ulf H Lerner
- From the Department of Molecular Periodontology, Umeå University, 90187 Umeå, Sweden, the Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition at Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, 413 45 Gothenburg, Sweden,
| |
Collapse
|
94
|
Wang G, Zhang X, Yu B, Ren K. Gliotoxin potentiates osteoblast differentiation by inhibiting nuclear factor-κB signaling. Mol Med Rep 2015; 12:877-84. [PMID: 25816130 PMCID: PMC4438943 DOI: 10.3892/mmr.2015.3524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 01/30/2015] [Indexed: 11/19/2022] Open
Abstract
The differentiation of pluripotent mesenchymal stem cells to mature osteoblasts is crucial for the maintenance of the adult skeleton. In rheumatic arthritis, osteoblast differentiation is impaired by the overproduction of cytokine tumor necrosis factor (TNF)-α. It has been demonstrated that TNF-α is able to inhibit osteoblast differentiation through the activation of nuclear factor (NF)-κB signaling. As a result of the critical role of TNF-α and NF-κB in the pathogenesis of bone-loss associated diseases, these factors are regarded as key targets for the development of therapeutic agents. In the current study, the role of the NF-κB inhibitor gliotoxin (GTX) in the regulation of osteoblast differentiation was evaluated. The non-toxic GTX doses were determined to be ≤3 μg/ml. It was revealed that GTX was able to block TNF-α-induced inhibition of osteoblast differentiation, as indicated by alkaline phosphatase (ALP) activity and ALP staining assays, as well as the expression levels of osteoblast-associated genes Col I, Ocn, Bsp, Runx2, Osx and ATF4. Additionally, it was identified that gliotoxin directly promoted bone morphoge-netic protein-2-induced osteoblast differentiation. GTX was found to inhibit the accumulation of NF-κB protein p65 in the nucleus and reduce NF-κB transcriptional activity, suggesting that GTX potentiated osteoblast differentiation via the suppression of NF-κB signaling.
Collapse
Affiliation(s)
- Guangye Wang
- The Third Department of Orthopaedics, Wuhu Second People's Hospital, Wuhu, Anhui 241000, P.R. China
| | - Xiaohai Zhang
- The Third Department of Orthopaedics, Wuhu Second People's Hospital, Wuhu, Anhui 241000, P.R. China
| | - Baoqing Yu
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, Shanghai 201399, P.R. China
| | - Ke Ren
- Department of Orthopedics, Nanjing General Hospital of Nanjing Military Command Region, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
95
|
Varley I, Hughes DC, Greeves JP, Stellingwerff T, Ranson C, Fraser WD, Sale C. RANK/RANKL/OPG pathway: genetic associations with stress fracture period prevalence in elite athletes. Bone 2015; 71:131-6. [PMID: 25464125 DOI: 10.1016/j.bone.2014.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 09/09/2014] [Accepted: 10/09/2014] [Indexed: 01/13/2023]
Abstract
CONTEXT The RANK/RANKL/OPG signalling pathway is important in the regulation of bone turnover, with single nucleotide polymorphisms (SNPs) in genes within this pathway associated with bone phenotypic adaptations. OBJECTIVE To determine whether four SNPs associated with genes in the RANK/RANKL/OPG signalling pathway were associated with stress fracture injury in elite athletes. DESIGN, PARTICIPANTS, AND METHODS Radiologically confirmed stress fracture history was reported in 518 elite athletes, forming the Stress Fracture Elite Athlete (SFEA) cohort. Data were analysed for the whole group and were sub-stratified into male and cases of multiple stress fracture groups. Genotypes were determined using proprietary fluorescence-based competitive allele-specific PCR assays. RESULTS SNPs rs3018362 (RANK) and rs1021188 (RANKL) were associated with stress fracture injury (P<0.05). 8.1% of the stress fracture group and 2.8% of the non-stress fracture group were homozygote for the rare allele of rs1021188. Allele frequency, heterozygotes and homozygotes for the rare allele of rs3018362 were associated with stress fracture period prevalence (P<0.05). Analysis of the male only group showed 8.2% of rs1021188 rare allele homozygotes had suffered a stress fracture whilst 2.5% of the non-stress fracture group were homozygous. In cases of multiple stress fractures, homozygotes for the rare allele of rs1021188 and individuals possessing at least one copy of the rare allele of rs4355801 (OPG) were shown to be associated with stress fracture injury (P<0.05). CONCLUSIONS The data support an association between SNPs in the RANK/RANKL/OPG signalling pathway and the development of stress fracture injury. The association of rs3018362 (RANK) and rs1021188 (RANKL) with stress fracture injury susceptibility supports their role in the maintenance of bone health and offers potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Ian Varley
- Biomedical Life and Health Sciences Research Centre, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK.
| | - David C Hughes
- Biomedical Life and Health Sciences Research Centre, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK.
| | - Julie P Greeves
- Department of Occupational Medicine, Headquarters Army Recruiting and Training Division, UK.
| | | | - Craig Ranson
- Cardiff School of Sport, Cardiff Metropolitan University, Cardiff, Wales, UK.
| | | | - Craig Sale
- Biomedical Life and Health Sciences Research Centre, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK.
| |
Collapse
|
96
|
Derer A, Böhm C, Grötsch B, Grün JR, Grützkau A, Stock M, Böhm S, Sehnert B, Gaipl U, Schett G, Hueber AJ, David JP. Rsk2 controls synovial fibroblast hyperplasia and the course of arthritis. Ann Rheum Dis 2014; 75:413-21. [DOI: 10.1136/annrheumdis-2014-205618] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 11/01/2014] [Indexed: 11/03/2022]
|
97
|
The aryl hydrocarbon receptor suppresses osteoblast proliferation and differentiation through the activation of the ERK signaling pathway. Toxicol Appl Pharmacol 2014; 280:502-10. [DOI: 10.1016/j.taap.2014.08.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 08/18/2014] [Accepted: 08/22/2014] [Indexed: 01/06/2023]
|
98
|
Bindarit, an inhibitor of monocyte chemotactic protein synthesis, protects against bone loss induced by chikungunya virus infection. J Virol 2014; 89:581-93. [PMID: 25339772 DOI: 10.1128/jvi.02034-14] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The recent global resurgence of arthritogenic alphaviruses, in particular chikungunya virus (CHIKV), highlights an urgent need for the development of therapeutic intervention strategies. While there has been significant progress in defining the pathophysiology of alphaviral disease, relatively little is known about the mechanisms involved in CHIKV-induced arthritis or potential therapeutic options to treat the severe arthritic symptoms associated with infection. Here, we used microcomputed tomographic (μCT) and histomorphometric analyses to provide previously undescribed evidence of reduced bone volume in the proximal tibial epiphysis of CHIKV-infected mice compared to the results for mock controls. This was associated with a significant increase in the receptor activator of nuclear factor-κB ligand/osteoprotegerin (RANKL/OPG) ratio in infected murine joints and in the serum of CHIKV patients. The expression levels of the monocyte chemoattractant proteins (MCPs), including MCP-1/CCL2, MCP-2/CCL8, and MCP-3/CCL7, were also highly elevated in joints of CHIKV-infected mice, accompanied by increased cellularity within the bone marrow in tibial epiphysis and ankle joints. Both this effect and CHIKV-induced bone loss were significantly reduced by treatment with the MCP inhibitor bindarit. Collectively, these findings demonstrate a unique role for MCPs in promoting CHIKV-induced osteoclastogenesis and bone loss during disease and suggest that inhibition of MCPs with bindarit may be an effective therapy for patients affected with alphavirus-induced bone loss. IMPORTANCE Arthritogenic alphaviruses, including chikungunya virus (CHIKV) and Ross River virus (RRV), cause worldwide outbreaks of polyarthritis, which can persist in patients for months following infection. Previous studies have shown that host proinflammatory soluble factors are associated with CHIKV disease severity. Furthermore, it is established that chemokine (C-C motif) ligand 2 (CCL2/MCP-1) is important in cellular recruitment and inducing bone-resorbing osteoclast (OC) formation. Here, we show that CHIKV replicates in bone and triggers bone loss by increasing the RANKL/OPG ratio. CHIKV infection results in MCP-induced cellular infiltration in the inflamed joints, and bone loss can be ameliorated by treatment with an MCP-inhibiting drug, bindarit. Taken together, our data reveal a previously undescribed role for MCPs in CHIKV-induced bone loss: one of recruiting monocytes/OC precursors to joint sites and thereby favoring a pro-osteoclastic microenvironment. This suggests that bindarit may be an effective treatment for alphavirus-induced bone loss and arthritis in humans.
Collapse
|
99
|
Exaggerated inflammatory environment decreases BMP-2/ACS-induced ectopic bone mass in a rat model: implications for clinical use of BMP-2. Osteoarthritis Cartilage 2014; 22:1186-96. [PMID: 24981632 DOI: 10.1016/j.joca.2014.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 06/09/2014] [Accepted: 06/13/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Numerous recent reports have observed a low osteoinductive efficacy property of bone morphogenetic protein-2 (BMP-2) and disappointing long-term outcomes in clinical cases. An alternative hypothesis, that these observations are caused by an exaggerated inflammatory environment, needs experimental evidence. METHOD Thirty-seven Sprague Dawley (SD) rats were administrated with Lipopolysaccharide (LPS) injections and BMP-2/absorbable collagen sponge (ACS) implantation to respectively mimic pre-operative and post-operative inflammatory responses. Blood samples and BMP-2/ACS implants were analyzed by enzyme-linked immunosorbent assay (ELISA), real-time polymerase chain reaction (PCR), micro-computed tomography (μCT) and histological examination. RESULTS LPS injections and BMP-2/ACS implantation provoked a significant elevation of inflammatory cytokines in serum and an obvious infiltration of inflammatory cells around BMP-2/ACS implants. The bone volume, mineral content and mineral density of the BMP-2/ACS implants from LPS-injected rats were significantly decreased, indicating that attenuated BMP-2-induced bone mass might be associated with down-regulated bone formation activity and up-regulated bone resorption activity. Furthermore, histological examination of the rhBMP-2/ACS implants showed a decreased expression of osteocalcin (OCN) and an increased number of osteoclasts in LPS-injected rats at 8 weeks; the expression level of bone turnover markers in serum and BMP-2/ACS implants revealed inhibited osteoblastogenesis activity and activated osteoclastogenesis activity in LPS-injected rats. Among the top three elevated pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) showed a suppressive effect on BMP-2-induced osteoblastic differentiation in vitro. CONCLUSION These data indicate that an exaggerated inflammatory environment may decrease BMP-2/ACS-induced bone mass in vivo by suppressing BMP-2-induced osteoblastic differentiation and by increasing the number or activity of osteoclasts. The negative role of exaggerated inflammation deserves consideration for future clinical use of BMP-2 in inducing bone regeneration.
Collapse
|
100
|
Klaus HD. Frontiers in the bioarchaeology of stress and disease: Cross-disciplinary perspectives from pathophysiology, human biology, and epidemiology. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2014; 155:294-308. [DOI: 10.1002/ajpa.22574] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 01/18/2023]
Affiliation(s)
- Haagen D. Klaus
- Department of Sociology and Anthropology; George Mason University; Fairfax VA
- Museo Nacional Sicán; Ferreñafe Peru
- Museo Nacional de Arqueología y Etnografía Hans Heinrich Brüning de Lambayeque; Lambayeque Peru
| |
Collapse
|