101
|
Okamoto K, Nakashima T, Shinohara M, Negishi-Koga T, Komatsu N, Terashima A, Sawa S, Nitta T, Takayanagi H. Osteoimmunology: The Conceptual Framework Unifying the Immune and Skeletal Systems. Physiol Rev 2017; 97:1295-1349. [DOI: 10.1152/physrev.00036.2016] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.
Collapse
Affiliation(s)
- Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Tomoki Nakashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Masahiro Shinohara
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takako Negishi-Koga
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Noriko Komatsu
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Asuka Terashima
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Shinichiro Sawa
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takeshi Nitta
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| |
Collapse
|
102
|
Collins FL, Rios-Arce ND, McCabe LR, Parameswaran N. Cytokine and hormonal regulation of bone marrow immune cell Wnt10b expression. PLoS One 2017; 12:e0181979. [PMID: 28800644 PMCID: PMC5553813 DOI: 10.1371/journal.pone.0181979] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/10/2017] [Indexed: 11/28/2022] Open
Abstract
Background & aims Wnt10b is a crucial regulator of bone density through its ability to promote osteoblastogenesis. Parathyroid hormone has been shown to regulate Wnt10b expression in CD8+ T cells. However, the relative expression and other source(s) of Wnt10b in the bone marrow immune cells (BMICs) is unknown. Sex hormones and cytokines such as, estrogen and TNFα are critical regulators of bone physiology but whether they regulate BMIC Wnt10b expression is unclear. To determine the potential regulation of Wnt10b by estrogen and TNFα, we assessed Wnt10b expression by flow cytometry under estrogen- and TNFα-deficient conditions. Methods Effects of TNFα was determined in male and female C57BL/6 wildtype and TNFα knockout mice. Effect of estrogen was investigated 4, 6 and 8 weeks post-surgery in ovariectomized Balb/c mice. Intracellular Wnt10b was detected using goat anti-mouse Wnt10b and a conjugated secondary antibody and analyzed by flow cytometry. Results Wnt10b expression was sex- and lineage-specific. Females had 1.8-fold higher Wnt10b signal compared to males. Percent of Wnt10b+ myeloid cells was higher in females than males (8.9% Vs 5.4%) but Wnt10b+ lymphoid cells was higher in males than females (6.3% Vs 2.5%). TNFα ablation in males increased total BM Wnt10b expression 1.5-fold but significantly reduced the percentage of BM Wnt10b+ CD4+ T cells (65%), CD8+ T cells (59%), dendritic cells (59%), macrophages (56%) and granulocytes (52%). These effects of TNFα on Wnt10b were observed only in males. In contrast to TNFα, estrogen-deficiency had indirect effects on BMIC Wnt10b levels; reducing the average percentage of BM Wnt10b+ CD8+ T cells (25%) and granulocytes (26%) across an 8-week time course. Conclusion Our results demonstrate unique cell type- and sex-dependent effects on BMIC Wnt10b expression. Together, our results reveal myeloid cells in the bone marrow as an important source of Wnt10b under complex hormonal and cytokine regulation.
Collapse
Affiliation(s)
- Fraser L. Collins
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
| | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
| | - Laura R. McCabe
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
- Department of Radiology, Michigan State University, East Lansing, Michigan, United States of America
- Biomedical Imaging Research Centre, Michigan State University, East Lansing, Michigan, United States of America
- * E-mail: (NP); (LRM)
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States of America
- * E-mail: (NP); (LRM)
| |
Collapse
|
103
|
Tsai YC, Yang BC, Peng WH, Lee YM, Yen MH, Cheng PY. Heme oxygenase-1 mediates anti-adipogenesis effect of raspberry ketone in 3T3-L1 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 31:11-17. [PMID: 28606512 DOI: 10.1016/j.phymed.2017.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/14/2017] [Accepted: 05/21/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Obesity is caused by excessive accumulation of body fat and is closely related to complex metabolic diseases. Raspberry ketone (RK), a major aromatic compound in red raspberry, was recently reported to possess anti-obesity effects. However, its mechanisms are unclear. AIM Adipogenesis plays a critical role in obesity and, therefore, this study aimed to investigate the effect and mechanisms of action of RK on adipogenesis in 3T3-L1 preadipocytes. MATERIALS AND METHODS 3T3-L1 preadipocytes were differentiated in medium containing insulin, dexamethasone, and 1-methyl-3-isobutylxanthine. Adipocyte lipid contents were determined using oil-red O staining while adipogenic transcription factor and lipogenic protein expressions were determined using western blotting. RESULTS RK (300-400µM) strongly inhibited lipid accumulation during 3T3-L1 preadipocyte differentiation into adipocytes. RK reduced the CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferation-activated receptor-γ (PPAR-γ), fatty acid synthase (FAS), and fatty acid-binding protein 4 (FABP4) expressions and increased heme oxygenase-1 (HO-1), Wnt10b, and β-catenin expressions in 3T3-L1 adipocytes. Additionally, RK inhibited lipid accumulation, and adipogenic transcription factor and lipogenic protein expressions were all decreased by inhibiting HO-1 or β-catenin using tin protoporphyrin (SnPP) or β-catenin short-interfering RNA (siRNA), respectively. Furthermore, Wnt10b and β-catenin expressions were negatively regulation by SnPP. CONCLUSION RK may exert anti-adipogenic effects through modulation of the HO-1/Wnt/beta-catenin signaling pathway.
Collapse
Affiliation(s)
- Yung-Chieh Tsai
- Department of Obstetrics and Gynecology, Chi-Mei Medical Center, Tainan; Department of Medicine, Taipei Medical University, Taipei; Department of Sport Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Bo-Cheng Yang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Wen-Huang Peng
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Yen-Mei Lee
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Mao-Hsiung Yen
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Pao-Yun Cheng
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
104
|
Zhu S, He H, Zhang C, Wang H, Gao C, Yu X, He C. Effects of pulsed electromagnetic fields on postmenopausal osteoporosis. Bioelectromagnetics 2017; 38:406-424. [PMID: 28665487 DOI: 10.1002/bem.22065] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 06/05/2017] [Indexed: 02/05/2023]
Abstract
Postmenopausal osteoporosis (PMOP) is considered to be a well-defined subject that has caused high morbidity and mortality. In elderly women diagnosed with PMOP, low bone mass and fragile bone strength have been proven to significantly increase risk of fragility fractures. Currently, various anabolic and anti-resorptive therapies have been employed in an attempt to retain healthy bone mass and strength. Pulsed electromagnetic fields (PEMFs), first applied in treating patients with delayed fracture healing and nonunions, may turn out to be another potential and effective therapy for PMOP. PEMFs can enhance osteoblastogenesis and inhibit osteoclastogenesis, thus contributing to an increase in bone mass and strength. However, accurate mechanisms of the positive effects of PEMFs on PMOP remain to be further elucidated. This review attempts to summarize recent advances of PEMFs in treating PMOP based on clinical trials, and animal and cellular studies. Possible mechanisms are also introduced, and the future possibility of application of PEMFs on PMOP are further explored and discussed. Bioelectromagnetics. 38:406-424, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Siyi Zhu
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Hongchen He
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Chi Zhang
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Haiming Wang
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Chengfei Gao
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Chengqi He
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P. R. China
- Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
105
|
Wang Q, Liu H, Wang Q, Zhou F, Liu Y, Zhang Y, Ding H, Yuan M, Li F, Chen Y. Involvement of c-Fos in cell proliferation, migration, and invasion in osteosarcoma cells accompanied by altered expression of Wnt2 and Fzd9. PLoS One 2017; 12:e0180558. [PMID: 28665975 PMCID: PMC5493424 DOI: 10.1371/journal.pone.0180558] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/16/2017] [Indexed: 01/04/2023] Open
Abstract
Osteosarcoma (OS) is an aggressive bone tumor, and proto-oncogene c-Fos is involved in this lethal disease. However, the role and molecular mechanism of c-Fos in the development and progression of OS remain enigmatic. As one of the Wnt family members, Wnt2 is closely associated with the development of several malignant tumors. In the present study, the expression of c-Fos, Wnt2, and its receptor Fzd9 in human OS tissues, MG63 OS cell line, and human osteoblast hFOB 1.19 cell line was detected by Western blot analysis, immunohistochemical staining, or reverse transcription-polymerase chain reaction. The role of c-Fos in the OS was clarified by treating MG63 cells with small interfering RNA to knockdown c-Fos. Then, cell migration and invasion were assayed by transwell assays and wound healing assay; cell proliferation was assayed by MTS method and 5-ethynyl-2'-deoxyuridine DNA proliferation in vitro detection; cell apoptosis was assayed by flow cytometric method. Co-immunoprecipitation kit was used to confirm the relationship between c-Fos and Wnt2/Fzd9. We found that the expression of c-Fos, Wnt2, and Fzd9 protein was distinctly higher in human OS tissues than that in the adjacent non-cancerous tissues, and their expression in the MG63 OS cell line was markedly increased compared with that in the human osteoblast hFOB 1.19 cell line. Knockdown of c-Fos inhibited the proliferation, migration, and invasion of MG63 cells, and promoted the apoptosis of MG63 cells. Moreover, knockdown of c-Fos inhibited the expression of Wnt2 and Fzd9 mRNA and protein. Our data enforced the evidence that knockdown of c-Fos inhibited cell proliferation, migration, and invasion, and promoted the apoptosis of OS cells accompanied by altered expression of Wnt2 and Fzd9. These findings offer new clues for OS development and progression, and c-Fos may be a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Qiaozhen Wang
- Department of Human Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Huancai Liu
- Affiliated hospital, Weifang Medical University, Weifang, Shandong, China
| | - Qing Wang
- Department of Human Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Fenghua Zhou
- Department of Pathology, Weifang Medical University, Weifang, Shandong, China
| | - Yongxin Liu
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Yawen Zhang
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Haoyu Ding
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Meng Yuan
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Fengjie Li
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Yanchun Chen
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
- * E-mail:
| |
Collapse
|
106
|
Joeng KS, Lee YC, Lim J, Chen Y, Jiang MM, Munivez E, Ambrose C, Lee BH. Osteocyte-specific WNT1 regulates osteoblast function during bone homeostasis. J Clin Invest 2017. [PMID: 28628032 DOI: 10.1172/jci92617] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mutations in WNT1 cause osteogenesis imperfecta (OI) and early-onset osteoporosis, identifying it as a key Wnt ligand in human bone homeostasis. However, how and where WNT1 acts in bone are unclear. To address this mechanism, we generated late-osteoblast-specific and osteocyte-specific WNT1 loss- and gain-of-function mouse models. Deletion of Wnt1 in osteocytes resulted in low bone mass with spontaneous fractures similar to that observed in OI patients. Conversely, Wnt1 overexpression from osteocytes stimulated bone formation by increasing osteoblast number and activity, which was due in part to activation of mTORC1 signaling. While antiresorptive therapy is the mainstay of OI treatment, it has limited efficacy in WNT1-related OI. In this study, anti-sclerostin antibody (Scl-Ab) treatment effectively improved bone mass and dramatically decreased fracture rate in swaying mice, a model of global Wnt1 loss. Collectively, our data suggest that WNT1-related OI and osteoporosis are caused in part by decreased mTORC1-dependent osteoblast function resulting from loss of WNT1 signaling in osteocytes. As such, this work identifies an anabolic function of osteocytes as a source of Wnt in bone development and homoeostasis, complementing their known function as targets of Wnt signaling in regulating osteoclastogenesis. Finally, this study suggests that Scl-Ab is an effective genotype-specific treatment option for WNT1-related OI and osteoporosis.
Collapse
Affiliation(s)
- Kyu Sang Joeng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yi-Chien Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Elda Munivez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Catherine Ambrose
- Department of Orthopedic Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
107
|
Fairfield H, Rosen CJ, Reagan MR. Connecting Bone and Fat: The Potential Role for Sclerostin. CURRENT MOLECULAR BIOLOGY REPORTS 2017; 3:114-121. [PMID: 28580233 PMCID: PMC5448707 DOI: 10.1007/s40610-017-0057-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sclerostin (SOST), a protein secreted from mature osteocytes in response to mechanical unloading and other stimuli, inhibits the osteogenic Wnt/β-catenin pathway in mesenchymal stem cells (MSCs) impeding their ability to differentiate into mineralizing osteoblasts. PURPOSE This review summarizes the crosstalk between adipose tissue and bone. It also reviews the origin, regulation, and role of SOST in osteogenesis and brings attention to an emerging role of this protein in the regulation of adipogenesis. RECENT FINDINGS Bone-derived molecules that drive MSC adipogenesis have not previously been identified, but recent findings suggest that SOST signaling may induce adipogenesis. In vivo SOST acts locally to induce changes in bone and, in vitro, increases adipogenesis in 3T3-L1 preadipocytes. SUMMARY SOST is able to induce adipogenesis in certain preadipocytes, however bone-specific studies are needed to determine the effect of local SOST concentrations in healthy and disease models on bone marrow adipose tissue.
Collapse
Affiliation(s)
- Heather Fairfield
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
| | - Clifford J. Rosen
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
| | - Michaela R. Reagan
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
| |
Collapse
|
108
|
Kawazoe M, Kaneko K, Shikano K, Kusunoki N, Nanki T, Kawai S. Glucocorticoid therapy causes contradictory changes of serum Wnt signaling-related molecules in systemic autoimmune diseases. Clin Rheumatol 2017; 37:2169-2178. [PMID: 28551822 DOI: 10.1007/s10067-017-3689-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/22/2022]
Abstract
The objective of this study was to investigate the clinical significance of the Wnt/β-catenin signaling pathway in glucocorticoid-induced osteoporosis. A total of 91 patients with systemic autoimmune diseases who received initial glucocorticoid therapy with prednisolone (30-60 mg daily) were prospectively enrolled. We measured serum levels of N-terminal peptide of type I procollagen (P1NP), bone alkaline phosphatase (BAP), tartrate-resistant acid phosphatase isoform 5b (TRACP-5b), N-telopeptide cross-linked type I collagen (NTX), sclerostin, Dickkopf-1 (Dkk-1), and Wnt3a before starting glucocorticoid therapy and every week for 4 weeks after its initiation. The effects of dexamethasone on expression of mRNA and protein of sclerostin and Dkk-1 by cultured normal human osteoblasts (NHOst) were evaluated by RT-PCR and ELISA, respectively. Serum levels of sclerostin and Dkk-1 increased significantly by 1 week of glucocorticoid therapy and then decreased from the second week onward. Serum Wnt3a tended to decrease and serum P1NP showed a significant decrease. However, TRACP-5b was significantly elevated from the first week of treatment onwards. In vitro study, dexamethasone increased Dkk-1 mRNA expression in cultured NHOst, but sclerostin mRNA was not detected. Dexamethasone also increased Dkk-1 protein production by osteoblasts, whereas sclerostin protein was not detected. Bone formation might be impaired at least in the first week of the initiation of glucocorticoid therapy by increase of the serum Wnt signaling inhibitors; however, their reductions in the subsequent weeks were contradictory to the maintained suppression of the bone formation markers after glucocorticoid therapy for patients with systemic autoimmune diseases.
Collapse
Affiliation(s)
- Mai Kawazoe
- Department of Internal Medicine, Graduate School of Medicine, Toho University, Tokyo, Japan.,Division of Rheumatology, Department of Internal Medicine, School of Medicine, Toho University, Tokyo, Japan
| | - Kaichi Kaneko
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Toho University, Tokyo, Japan
| | - Kotaro Shikano
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Toho University, Tokyo, Japan
| | - Natsuko Kusunoki
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Toho University, Tokyo, Japan
| | - Toshihiro Nanki
- Department of Internal Medicine, Graduate School of Medicine, Toho University, Tokyo, Japan.,Division of Rheumatology, Department of Internal Medicine, School of Medicine, Toho University, Tokyo, Japan
| | - Shinichi Kawai
- Department of Inflammation and Pain Control Research, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ota-ku, Tokyo, 143-8540, Japan.
| |
Collapse
|
109
|
Zhou J, Wang S, Qi Q, Yang X, Zhu E, Yuan H, Li X, Liu Y, Li X, Wang B. Nuclear factor I-C reciprocally regulates adipocyte and osteoblast differentiation via control of canonical Wnt signaling. FASEB J 2017; 31:1939-1952. [PMID: 28122918 DOI: 10.1096/fj.201600975rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/09/2017] [Indexed: 11/20/2024]
Abstract
Nuclear factor I-C (NFIC) has recently been identified as an important player in osteogenesis and bone homeostasis in vivo However, the molecular mechanisms involved have yet to be defined. In the current study, Nfic expression was altered in primary marrow stromal cells and established progenitor lines after adipogenic and osteogenic treatment. Overexpression of Nfic in stromal cells ST2, mesenchymal cells C3H10T1/2, and primary marrow stromal cells inhibited adipogenic differentiation, whereas it promoted osteogenic differentiation. Conversely, silencing of endogenous Nfic in the cell lines enhanced adipogenic differentiation, whereas it blocked osteogenic differentiation. Mechanism investigations revealed that Nfic overexpression promoted nuclear translocation of β-catenin and increased nuclear protein levels of β-catenin and transcription factor 7-like 2 (TCF7L2). Promoter studies and the chromatin immunoprecipitation (ChIP) assay revealed that NFIC directly binds to the promoter of low-density lipoprotein receptor-related protein 5 (Lrp5) and thereafter transactivates the promoter. Finally, inactivation of canonical Wnt signaling in ST2 attenuated the inhibition of adipogenic differentiation and stimulation of osteogenic differentiation by NFIC. Our study suggests that NFIC balances adipogenic and osteogenic differentiation from progenitor cells through controlling canonical Wnt signaling and highlights the potential of NFIC as a target for new therapies to control metabolic disorders like osteoporosis and obesity.-Zhou, J., Wang, S., Qi, Q., Yang, X., Zhu, E., Yuan, H., Li, X., Liu, Y., Li, X., Wang, B. Nuclear factor I-C reciprocally regulates adipocyte and osteoblast differentiation via control of canonical Wnt signaling.
Collapse
Affiliation(s)
- Jie Zhou
- Key Laboratory of Hormones and Development, Ministry of Health, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- 2011 Collaborative Innovation Center for Metabolic Diseases, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | - Shan Wang
- College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; and
| | - Qi Qi
- Key Laboratory of Hormones and Development, Ministry of Health, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaoyue Yang
- Stomatological Hospital, Tianjin Medical University, Tianjin, China
| | - Endong Zhu
- Key Laboratory of Hormones and Development, Ministry of Health, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Hairui Yuan
- Key Laboratory of Hormones and Development, Ministry of Health, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xuemei Li
- Key Laboratory of Hormones and Development, Ministry of Health, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Ying Liu
- Stomatological Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaoxia Li
- College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; and
| | - Baoli Wang
- Key Laboratory of Hormones and Development, Ministry of Health, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China;
- 2011 Collaborative Innovation Center for Metabolic Diseases, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
110
|
Zhou W, Huang H, Zhu H, Zhou P, Shi X. New metabolites from the biotransformation of ginsenoside Rb1 by Paecilomyces bainier sp.229 and activities in inducing osteogenic differentiation by Wnt/β-catenin signaling activation. J Ginseng Res 2017; 42:199-207. [PMID: 29719467 PMCID: PMC5925613 DOI: 10.1016/j.jgr.2017.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/16/2017] [Indexed: 10/25/2022] Open
Abstract
Background Ginseng is a well-known traditional Chinese medicine that has been widely used in a range of therapeutic and healthcare applications in East Asian countries. Microbial transformation is regarded as an effective and useful technology in modification of nature products for finding new chemical derivatives with potent bioactivities. In this study, three minor derivatives of ginsenoside compound K were isolated and the inducing effects in the Wingless-type MMTV integration site (Wnt) signaling pathway were also investigated. Methods New compounds were purified from scale-up fermentation of ginsenoside Rb1 by Paecilomyces bainier sp. 229 through repeated silica gel column chromatography and high pressure liquid chromatography. Their structures were determined based on spectral data and X-ray diffraction. The inductive activities of these compounds on the Wnt signaling pathway were conducted on MC3T3-E1 cells by quantitative real-time polymerase chain reaction analysis. Results The structures of a known 3-keto derivative and two new dehydrogenated metabolites were elucidated. The crystal structure of the 3-keto derivative was reported for the first time and its conformation was compared with that of ginsenoside compound K. The inductive effects of these compounds on osteogenic differentiation by activating the Wnt/β-catenin signaling pathway were explained for the first time. Conclusion This study may provide a new insight into the metabolic pathway of ginsenoside by microbial transformation. In addition, the results might provide a reasonable explanation for the activity of ginseng in treating osteoporosis and supply good monomer ginsenoside resources for nutraceutical or pharmaceutical development.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Chemistry, Fudan University, Shanghai, China
| | - Hai Huang
- School of Pharmacy, Fudan University, Shanghai, China
| | - Haiyan Zhu
- School of Pharmacy, Fudan University, Shanghai, China
| | - Pei Zhou
- School of Pharmacy, Fudan University, Shanghai, China
| | - Xunlong Shi
- School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
111
|
Alteration of osteoblast arrangement via direct attack by cancer cells: New insights into bone metastasis. Sci Rep 2017; 7:44824. [PMID: 28303941 PMCID: PMC5356003 DOI: 10.1038/srep44824] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/15/2017] [Indexed: 11/09/2022] Open
Abstract
Intact bone tissue exhibits a characteristic anisotropic microstructure derived from collagen fiber alignment and the related c-axis orientation of apatite crystals, which govern the mechanical properties of bone tissue. In contrast, tumor-invaded bone exhibits a disorganized, less-aligned microstructure that results in severely disrupted mechanical function. Despite its importance both in basic principle and in therapeutic applications, the classical understanding of bone metastasis is limited to alterations in bone mass regulated by metastatic cancer cells. In this study, we demonstrate a novel mechanism underlying the disruption of bone tissue anisotropy in metastasized bone. We observed that direct attack by cancer cells on osteoblasts induces the less-organized osteoblast arrangement. Importantly, the crystallographic anisotropy of bone tissue is quantitatively determined by the level of osteoblast arrangement. Osteoblast arrangement was significantly disrupted by physical contact with cancer cells such as osteolytic melanoma B16F10, breast cancer MDA-MB-231, and osteoblastic prostate cancer MDA-PCa-2b cells. The present findings demonstrate that the abnormal arrangement of osteoblasts induced by physical contact with cancer cells facilitates the disorganized microstructure of metastasized bone.
Collapse
|
112
|
Sulston RJ, Cawthorn WP. Bone marrow adipose tissue as an endocrine organ: close to the bone? Horm Mol Biol Clin Investig 2017; 28:21-38. [PMID: 27149203 DOI: 10.1515/hmbci-2016-0012] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
Abstract
White adipose tissue (WAT) is a major endocrine organ, secreting a diverse range of hormones, lipid species, cytokines and other factors to exert diverse local and systemic effects. These secreted products, known as 'adipokines', contribute extensively to WAT's impact on physiology and disease. Adipocytes also exist in the bone marrow (BM), but unlike WAT, study of this bone marrow adipose tissue (MAT) has been relatively limited. We recently discovered that MAT contributes to circulating adiponectin, an adipokine that mediates cardiometabolic benefits. Moreover, we found that MAT expansion exerts systemic effects. Together, these observations identify MAT as an endocrine organ. Additional studies are revealing further secretory functions of MAT, including production of other adipokines, cytokines and lipids that exert local effects within bone. These observations suggest that, like WAT, MAT has secretory functions with diverse potential effects, both locally and systemically. A major limitation is that these findings are often based on in vitro approaches that may not faithfully recapitulate the characteristics and functions of BM adipocytes in vivo. This underscores the need to develop improved methods for in vivo analysis of MAT function, including more robust transgenic models for MAT targeting, and continued development of techniques for non-invasive analysis of MAT quantity and quality in humans. Although many aspects of MAT formation and function remain poorly understood, MAT is now attracting increasing research focus; hence, there is much promise for further advances in our understanding of MAT as an endocrine organ, and how MAT impacts human health and disease.
Collapse
|
113
|
Siddiqui JA, Partridge NC. Physiological Bone Remodeling: Systemic Regulation and Growth Factor Involvement. Physiology (Bethesda) 2017; 31:233-45. [PMID: 27053737 DOI: 10.1152/physiol.00061.2014] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bone remodeling is essential for adult bone homeostasis. It comprises two phases: bone formation and resorption. The balance between the two phases is crucial for sustaining bone mass and systemic mineral homeostasis. This review highlights recent work on physiological bone remodeling and discusses our knowledge of how systemic and growth factors regulate this process.
Collapse
Affiliation(s)
- Jawed A Siddiqui
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Nicola C Partridge
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| |
Collapse
|
114
|
Bone and adipose tissue formation. Z Rheumatol 2017. [DOI: 10.1007/s00393-016-0143-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
115
|
Iyer S, Han L, Ambrogini E, Yavropoulou M, Fowlkes J, Manolagas SC, Almeida M. Deletion of FoxO1, 3, and 4 in Osteoblast Progenitors Attenuates the Loss of Cancellous Bone Mass in a Mouse Model of Type 1 Diabetes. J Bone Miner Res 2017; 32:60-69. [PMID: 27491024 PMCID: PMC5492385 DOI: 10.1002/jbmr.2934] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes is associated with osteopenia and increased fragility fractures, attributed to reduced bone formation. However, the molecular mechanisms mediating these effects remain unknown. Insulin promotes osteoblast formation and inhibits the activity of the FoxO transcription factors. FoxOs, on the other hand, inhibit osteoprogenitor proliferation and bone formation. Here, we investigated whether FoxOs play a role in the low bone mass associated with type 1 diabetes, using mice lacking FoxO1, 3, and 4 in osteoprogenitor cells (FoxO1,3,4ΔOsx1-Cre ). Streptozotocin-induced diabetes caused a reduction in bone mass and strength in FoxO-intact mice. In contrast, cancellous bone was unaffected in diabetic FoxO1,3,4ΔOsx1-Cre mice. The low bone mass in the FoxO-intact diabetic mice was associated with decreased osteoblast number and bone formation, as well as decreased expression of the anti-osteoclastogenic cytokine osteoprotegerin (OPG) and increased osteoclast number. FoxO deficiency did not alter the effects of diabetes on bone formation; however, it did prevent the decrease in OPG and the increase in osteoclast number. Addition of high glucose to osteoblastic cell cultures decreased OPG mRNA, indicating that hyperglycemia in and of itself contributes to diabetic bone loss. Taken together, these results suggest that FoxOs exacerbate the loss of cancellous bone mass associated with type 1 diabetes and that inactivation of FoxOs might ameliorate the adverse effects of insulin deficiency. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Srividhya Iyer
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Li Han
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Maria Yavropoulou
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - John Fowlkes
- Barnstable Brown Diabetes and Obesity Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences and the Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| |
Collapse
|
116
|
Chkourko Gusky H, Diedrich J, MacDougald OA, Podgorski I. Omentum and bone marrow: how adipocyte-rich organs create tumour microenvironments conducive for metastatic progression. Obes Rev 2016; 17:1015-1029. [PMID: 27432523 PMCID: PMC5056818 DOI: 10.1111/obr.12450] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/30/2022]
Abstract
A number of clinical studies have linked adiposity with increased cancer incidence, progression and metastasis, and adipose tissue is now being credited with both systemic and local effects on tumour development and survival. Adipocytes, a major component of benign adipose tissue, represent a significant source of lipids, cytokines and adipokines, and their presence in the tumour microenvironment substantially affects cellular trafficking, signalling and metabolism. Cancers that have a high predisposition to metastasize to the adipocyte-rich host organs are likely to be particularly affected by the presence of adipocytes. Although our understanding of how adipocytes influence tumour progression has grown significantly over the last several years, the mechanisms by which adipocytes regulate the metastatic niche are not well-understood. In this review, we focus on the omentum, a visceral white adipose tissue depot, and the bone, a depot for marrow adipose tissue, as two distinct adipocyte-rich organs that share common characteristic: they are both sites of significant metastatic growth. We highlight major differences in origin and function of each of these adipose depots and reveal potential common characteristics that make them environments that are attractive and conducive to secondary tumour growth. Special attention is given to how omental and marrow adipocytes modulate the tumour microenvironment by promoting angiogenesis, affecting immune cells and altering metabolism to support growth and survival of metastatic cancer cells.
Collapse
Affiliation(s)
- H Chkourko Gusky
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
| | - J Diedrich
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA.,Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - O A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - I Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA. .,Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
117
|
Liu YQ, Han XF, Bo JX, Ma HP. Wedelolactone Enhances Osteoblastogenesis but Inhibits Osteoclastogenesis through Sema3A/NRP1/PlexinA1 Pathway. Front Pharmacol 2016; 7:375. [PMID: 27803667 PMCID: PMC5067540 DOI: 10.3389/fphar.2016.00375] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/27/2016] [Indexed: 02/02/2023] Open
Abstract
Bone remodeling balance is maintained by tight coupling of osteoblast-mediated bone formation and osteoclast-mediated bone resorption. Thus, agents with the capacity to regulate osteoblastogenesis and osteoclastogenesis have been investigated for therapy of bone-related diseases such as osteoporosis. In this study, we found that wedelolactone, a compound isolated from Ecliptae herba, and a 9-day incubation fraction of conditioned media obtained from wedelolactone-treated bone marrow mesenchymal stem cell (BMSC) significantly inhibited tartrate-resistant acid phosphatase (TRAP) activity in RANKL-stimulated osteoclastic RAW264.7 cells. Addition of the semaphorin 3A (Sema3A) antibody to the conditioned media partially blocked the medium’s inhibitory effects on the RAW264.7 cells. In BMSC, mRNA expression of Sema3A increased in the presence of different wedelolactone concentrations. Blocking Sema3A activity with its antibody reversed wedelolactone-induced alkaline phosphatase activity in BMSC and concurrently enhanced wedelolactone-reduced TRAP activity in osteoclastic RAW264.7 cells. Moreover, in BMSC, wedelolactone enhanced binding of Sema3A with cell-surface receptors, including neuropilin (NRP)1 and plexinA1. Furthermore, nuclear accumulation of β-catenin, a transcription factor acting downstream of wedelolactone-induced Sema3A signaling, was blocked by the Sema3A antibody. In osteoclastic RAW264.7 cells, conditioned media and wedelolactone promoted the formation of plexin A1-NRP1, but conditioned media also caused the sequestration of the plexin A1-DNAX-activating protein 12 (DAP12) complex and suppressed the phosphorylation of phospholipase C (PLC)γ2. These data suggest that wedelolactone promoted osteoblastogenesis through production of Sema3A, thus inducing the formation of a Sema3A-plexinA1-Nrp1 complex and β-catenin activation. In osteoclastic RAW264.7 cells, wedelolactone inhibited osteoclastogenesis through sequestration of the plexinA1-DAP12 complex, induced the formation of plexinA1-Nrp1 complex, and suppressed PLCγ2 activation.
Collapse
Affiliation(s)
- Yan-Qiu Liu
- Institute (College) of Integrative Medicine, Dalian Medical University Dalian, China
| | - Xiao-Fei Han
- Glucose and Lipid Metabolism Laboratory of Liaoning Province, College of Life Science and Technology, Dalian University Dalian, China
| | - Jun-Xia Bo
- Glucose and Lipid Metabolism Laboratory of Liaoning Province, College of Life Science and Technology, Dalian University Dalian, China
| | - Hui-Peng Ma
- College of Medical Laboratory, Dalian Medical University Dalian, China
| |
Collapse
|
118
|
Fu S, Yang L, Hong H, Zhang R. [Wnt/β-catenin signaling is involved in the Icariin induced proliferation of bone marrow mesenchymal stem cells]. J TRADIT CHIN MED 2016; 36:360-8. [PMID: 27468552 DOI: 10.1016/s0254-6272(16)30050-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To investigate the effect of icariin on proliferation of bone marrow mesenchymal stem cells (BMSCs) in Sprague-Dawley (SD) rats. METHODS BMSCs were obtained from SD rat bone marrow with differential time adherent method. Its characteristic was identified through differentiation cell surface antigens and the multi-lineage (osteo/adipo/chondo) differentiation potential. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method and 5-Bromo-2-Deoxyuridine (BrdU) incorporation were applied to detect the effect of icariin on BMSCs proliferation. Flow cytometry was used to detect proliferation index of BMSCs. The RNA level and the distribution of β-catenin were evaluated by Real-time Polymerase Chain Reaction (PCR) and Immunofluorescent staining respectively. Western blot was used to detect protein expression levels of β-catenin, glycogen synthase kinase-3 beta (GSK-3β), phospho-glycogen synthase kinase-3 beta (pGSK-3β) and cyclinD1. RESULTS Icariin promoted BMSCs proliferation at the concentration of 0.05-2.0 mg/L. The percentage of BrdU positive cells of BMSCs was increased from 40.98% to 70.42%, and the proliferation index value was increased from 8.9% to 17.5% with the treatment of 0.05 mg/L icariin, which significance values were both less than 0.05. Compared with the control group, total and nuclear β-catenin proteins, as well as β-catenin mRNA expression, were all increased with icariin treatment. Meanwhile, the phosphorylation level of GSK-3β and cyclinD1 protein expressions were also increased in BMSCs with icariin treatment. CONCLUSION The findings of the present study demonstrated that low dosage of icariin could promote BMSCs proliferation. The activation of Wnt/β-catenin pathways was involved in this process.
Collapse
|
119
|
Abstract
Leptin has been described to have a crucial role in bone homeostasis by systemic as well as local action. Systemically, leptin seems to inhibit bone formation controlled by a feedback loop including osteocalcin and insulin. Even though the action seems to be bone site specific, as well as gender- and time-dependent, the results showing the interaction of these three factors are in part still inconsistent. In this article the complex effects of leptin, insulin, and osteocalcin on bone and fat metabolism are summarized.
Collapse
|
120
|
Phetfong J, Sanvoranart T, Nartprayut K, Nimsanor N, Seenprachawong K, Prachayasittikul V, Supokawej A. Osteoporosis: the current status of mesenchymal stem cell-based therapy. Cell Mol Biol Lett 2016; 21:12. [PMID: 28536615 PMCID: PMC5414670 DOI: 10.1186/s11658-016-0013-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/25/2016] [Indexed: 12/21/2022] Open
Abstract
Osteoporosis, or bone loss, is a progressive, systemic skeletal disease that affects millions of people worldwide. Osteoporosis is generally age related, and it is underdiagnosed because it remains asymptomatic for several years until the development of fractures that confine daily life activities, particularly in elderly people. Most patients with osteoporotic fractures become bedridden and are in a life-threatening state. The consequences of fracture can be devastating, leading to substantial morbidity and mortality of the patients. The normal physiologic process of bone remodeling involves a balance between bone resorption and bone formation during early adulthood. In osteoporosis, this process becomes imbalanced, resulting in gradual losses of bone mass and density due to enhanced bone resorption and/or inadequate bone formation. Several growth factors underlying age-related osteoporosis and their signaling pathways have been identified, such as osteoprotegerin (OPG)/receptor activator of nuclear factor B (RANK)/RANK ligand (RANKL), bone morphogenetic protein (BMP), wingless-type MMTV integration site family (Wnt) proteins and signaling through parathyroid hormone receptors. In addition, the pathogenesis of osteoporosis has been connected to genetics. The current treatment of osteoporosis predominantly consists of antiresorptive and anabolic agents; however, the serious adverse effects of using these drugs are of concern. Cell-based replacement therapy via the use of mesenchymal stem cells (MSCs) may become one of the strategies for osteoporosis treatment in the future.
Collapse
Affiliation(s)
- Jitrada Phetfong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Tanwarat Sanvoranart
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Kuneerat Nartprayut
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Natakarn Nimsanor
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Kanokwan Seenprachawong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Phuttamonthon, Salaya, Nakhon Pathom 73170 Thailand
| |
Collapse
|
121
|
Fu Y, Gao C, Liang Y, Wang M, Huang Y, Ma W, Li T, Jia Y, Yu F, Zhu W, Cui Q, Li Y, Xu Q, Wang X, Kong W. Shift of Macrophage Phenotype Due to Cartilage Oligomeric Matrix Protein Deficiency Drives Atherosclerotic Calcification. Circ Res 2016; 119:261-276. [PMID: 27151399 DOI: 10.1161/circresaha.115.308021] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 05/05/2016] [Indexed: 12/29/2022]
Abstract
RATIONALE Intimal calcification is highly correlated with atherosclerotic plaque burden, but the underlying mechanism is poorly understood. We recently reported that cartilage oligomeric matrix protein (COMP), a component of vascular extracellular matrix, is an endogenous inhibitor of vascular smooth muscle cell calcification. OBJECTIVE To investigate whether COMP affects atherosclerotic calcification. METHODS AND RESULTS ApoE(-/-)COMP(-/-) mice fed with chow diet for 12 months manifested more extensive atherosclerotic calcification in the innominate arteries than did ApoE(-/-) mice. To investigate which origins of COMP contributed to atherosclerotic calcification, bone marrow transplantation was performed between ApoE(-/-) and ApoE(-/-)COMP(-/-) mice. Enhanced calcification was observed in mice transplanted with ApoE(-/-)COMP(-/-) bone marrow compared with mice transplanted with ApoE(-/-) bone marrow, indicating that bone marrow-derived COMP may play a critical role in atherosclerotic calcification. Furthermore, microarray profiling of wild-type and COMP(-/-) macrophages revealed that COMP-deficient macrophages exerted atherogenic and osteogenic characters. Integrin β3 protein was attenuated in COMP(-/-) macrophages, and overexpression of integrin β3 inhibited the shift of macrophage phenotypes by COMP deficiency. Furthermore, adeno-associated virus 2-integrin β3 infection attenuated atherosclerotic calcification in ApoE(-/-)COMP(-/-) mice. Mechanistically, COMP bound directly to β-tail domain of integrin β3 via its C-terminus, and blocking of the COMP-integrin β3 association by β-tail domain mimicked the COMP deficiency-induced shift in macrophage phenotypes. Similar to COMP deficiency in mice, transduction of adeno-associated virus 2-β-tail domain enhanced atherosclerotic calcification in ApoE(-/-) mice. CONCLUSIONS These results reveal that COMP deficiency acted via integrin β3 to drive macrophages toward the atherogenic and osteogenic phenotype and thereby aggravate atherosclerotic calcification.
Collapse
Affiliation(s)
- Yi Fu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Cheng Gao
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Ying Liang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Meili Wang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yaqian Huang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Wei Ma
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Tuoyi Li
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yiting Jia
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Fang Yu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Wanlin Zhu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Qinghua Cui
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yanhui Li
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Qingbo Xu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Xian Wang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Wei Kong
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.).
| |
Collapse
|
122
|
Jiang M, Zheng C, Shou P, Li N, Cao G, Chen Q, Xu C, Du L, Yang Q, Cao J, Han Y, Li F, Cao W, Liu F, Rabson A, Roberts A, Xie W, Wang Y, Shi Y. SHP1 Regulates Bone Mass by Directing Mesenchymal Stem Cell Differentiation. Cell Rep 2016; 16:769-80. [DOI: 10.1016/j.celrep.2016.06.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 04/03/2016] [Accepted: 06/05/2016] [Indexed: 12/31/2022] Open
|
123
|
Scheller EL, Burr AA, MacDougald OA, Cawthorn WP. Inside out: Bone marrow adipose tissue as a source of circulating adiponectin. Adipocyte 2016; 5:251-69. [PMID: 27617171 PMCID: PMC5014002 DOI: 10.1080/21623945.2016.1149269] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 02/09/2023] Open
Abstract
The adipocyte-derived hormone adiponectin mediates beneficial cardiometabolic effects, and hypoadiponectinemia is a biomarker for increased metabolic and cardiovascular risk. Indeed, circulating adiponectin decreases in obesity and insulin-resistance, likely because of impaired production from white adipose tissue (WAT). Conversely, lean states such as caloric restriction (CR) are characterized by hyperadiponectinemia, even without increased adiponectin production from WAT. The reasons underlying this paradox have remained elusive, but our recent research suggests that CR-associated hyperadiponectinemia derives from an unexpected source: bone marrow adipose tissue (MAT). Herein, we elaborate on this surprising discovery, including further discussion of potential mechanisms influencing adiponectin production from MAT; additional evidence both for and against our conclusions; and observations suggesting that the relationship between MAT and adiponectin might extend beyond CR. While many questions remain, the burgeoning study of MAT promises to reveal further key insights into MAT biology, both as a source of adiponectin and beyond.
Collapse
|
124
|
Gautam J, Khedgikar V, Choudhary D, Kushwaha P, Dixit P, Singh D, Maurya R, Trivedi R. An isoflavone cladrin prevents high-fat diet-induced bone loss and inhibits the expression of adipogenic gene regulators in 3T3-L1 adipocyte. J Pharm Pharmacol 2016; 68:1051-63. [DOI: 10.1111/jphp.12562] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/29/2016] [Indexed: 01/05/2023]
Abstract
Abstract
Objective
This study evaluates the effect of isoflavone cladrin on high-fat diet (HFD)-induced bone loss and adipogenesis.
Methods
Thirty-two 4-week-old male C57BL/6J mice were divided into four groups: a standard diet group, a HFD group and HFD group with cladrin (5 and 10 mg/kg per day orally) for 12 weeks. The effect of cladrin on bone micro-architecture, bone marrow cell lineages and hyperlipidaemia were assessed. For assessing anti-adipogenic activity of cladrin, 3T3-L1 cells were used.
Key findings
Cladrin attenuated HFD-induced hyperlipidaemia and bone loss by preserving bone micro-architecture and strength. Effect of cladrin was found at the level of bone marrow progenitor cells. Gene expression profile of cladrin-treated mice bone showed upregulation of osteoblast and downregulation of adipogenic transcription factors and increased OPG/RANKL ratio. Cladrin inhibited cellular lipid accumulation through downregulation of transcription factors such as PPAR-γ and C/EBP-α and modulated the expression of major adipokines involved behind obesity stimulation without eliciting cell cytotoxicity in 3T3-L1 adipocytes.
Conclusion
We conclude that cladrin may improve obesity-induced bone loss and hyperlipidaemia in mice fed HFD and adipogenesis in 3T3-L1 cells by modifying adipokines and could offer clinical benefits as a supplement to treat obesity-induced disorders.
Collapse
Affiliation(s)
- Jyoti Gautam
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vikram Khedgikar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | | | - Priyanka Kushwaha
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Preeti Dixit
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Divya Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rakesh Maurya
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ritu Trivedi
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
125
|
Liu Y, Almeida M, Weinstein RS, O'Brien CA, Manolagas SC, Jilka RL. Skeletal inflammation and attenuation of Wnt signaling, Wnt ligand expression, and bone formation in atherosclerotic ApoE-null mice. Am J Physiol Endocrinol Metab 2016; 310:E762-73. [PMID: 26956187 PMCID: PMC6415649 DOI: 10.1152/ajpendo.00501.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/07/2016] [Indexed: 12/18/2022]
Abstract
ApoE-null (ApoE-KO) mice fed a high-fat diet (HFD) develop atherosclerosis, due in part to activation of vascular inflammation by oxidized low-density lipoprotein. Since bone loss also occurs in these mice, we used them to investigate the impact of oxidized lipids on bone homeostasis and to search for underlying pathogenic pathways. Four-month-old female ApoE-KO mice fed a HFD for three months exhibited increased levels of oxidized lipids in bone, as well as decreased femoral and vertebral trabecular and cortical bone mass, compared with ApoE-KO mice on normal diet. Despite HFD-induced increase in expression of Alox15, a lipoxygenase that oxidizes LDL and promotes atherogenesis, global deletion of this gene failed to ameliorate the skeletal impact of HFD. Osteoblast number and function were dramatically reduced in trabecular and cortical bone of HFD-fed mice, whereas osteoclast number was modestly reduced only in trabecular bone, indicating that an imbalance in favor of osteoclasts was responsible for HFD-induced bone loss. These changes were associated with decreased osteoblast progenitors and increased monocyte/macrophages in the bone marrow as well as increased expression of IL-1β, IL-6, and TNF. HFD also attenuated Wnt signaling as evidenced by reduced expression of Wnt target genes, and it decreased expression of pro-osteoblastogenic Wnt ligands. These results suggest that oxidized lipids decrease bone mass by increasing anti-osteoblastogenic inflammatory cytokines and decreasing pro-osteoblastogenic Wnt ligands.
Collapse
Affiliation(s)
- Yu Liu
- Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Maria Almeida
- Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Robert S Weinstein
- Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Charles A O'Brien
- Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Stavros C Manolagas
- Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Robert L Jilka
- Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
126
|
Cain CJ, Valencia JT, Ho S, Jordan K, Mattingly A, Morales BM, Hsiao EC. Increased Gs Signaling in Osteoblasts Reduces Bone Marrow and Whole-Body Adiposity in Male Mice. Endocrinology 2016; 157:1481-94. [PMID: 26901092 PMCID: PMC4816728 DOI: 10.1210/en.2015-1867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/16/2016] [Indexed: 12/21/2022]
Abstract
Bone is increasingly recognized as an endocrine organ that can regulate systemic hormones and metabolism through secreted factors. Although bone loss and increased adiposity appear to be linked clinically, whether conditions of increased bone formation can also change systemic metabolism remains unclear. In this study, we examined how increased osteogenesis affects metabolism by using an engineered G protein-coupled receptor, Rs1, to activate Gs signaling in osteoblastic cells in ColI(2.3)(+)/Rs1(+) transgenic mice. We previously showed that these mice have dramatically increased bone formation resembling fibrous dysplasia of the bone. We found that total body fat was significantly reduced starting at 3 weeks of age. Furthermore, ColI(2.3)(+)/Rs1(+) mice showed reduced O2 consumption and respiratory quotient measures without effects on food intake and energy expenditure. The mice had significantly decreased serum triacylglycerides, leptin, and adiponectin. Resting glucose and insulin levels were unchanged; however, glucose and insulin tolerance tests revealed increased sensitivity to insulin. The mice showed resistance to fat accumulation from a high-fat diet. Furthermore, ColI(2.3)(+)/Rs1(+) mouse bones had dramatically reduced mature adipocyte differentiation, increased Wingless/Int-1 (Wnt) signaling, and higher osteoblastic glucose utilization than controls. These findings suggest that osteoblasts can influence both local and peripheral adiposity in conditions of increased bone formation and suggest a role for osteoblasts in the regulation of whole-body adiposity and metabolic homeostasis.
Collapse
Affiliation(s)
- Corey J Cain
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Joel T Valencia
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Samantha Ho
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Kate Jordan
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Aaron Mattingly
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Blanca M Morales
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| | - Edward C Hsiao
- Department of Medicine, Division of Endocrinology and Metabolism; Institute for Human Genetics; and Program in Craniofacial Biology (C.J.C., S.H., K.J., A.M., B.M.M., and E.C.H.); and the Biomedical Sciences Graduate Program (J.T.V. and E.C.H.); University of California, San Francisco, San Francisco, California 94143-0794
| |
Collapse
|
127
|
Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ 2016; 23:1128-39. [PMID: 26868907 PMCID: PMC4946886 DOI: 10.1038/cdd.2015.168] [Citation(s) in RCA: 887] [Impact Index Per Article: 98.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 11/03/2015] [Accepted: 12/01/2015] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs), a non-hematopoietic stem cell population first discovered in bone marrow, are multipotent cells capable of differentiating into mature cells of several mesenchymal tissues, such as fat and bone. As common progenitor cells of adipocytes and osteoblasts, MSCs are delicately balanced for their differentiation commitment. Numerous in vitro investigations have demonstrated that fat-induction factors inhibit osteogenesis, and, conversely, bone-induction factors hinder adipogenesis. In fact, a variety of external cues contribute to the delicate balance of adipo-osteogenic differentiation of MSCs, including chemical, physical, and biological factors. These factors trigger different signaling pathways and activate various transcription factors that guide MSCs to commit to either lineage. The dysregulation of the adipo-osteogenic balance has been linked to several pathophysiologic processes, such as aging, obesity, osteopenia, osteopetrosis, and osteoporosis. Thus, the regulation of MSC differentiation has increasingly attracted great attention in recent years. Here, we review external factors and their signaling processes dictating the reciprocal regulation between adipocytes and osteoblasts during MSC differentiation and the ultimate control of the adipo-osteogenic balance.
Collapse
|
128
|
Jacobs FA, Sadie-Van Gijsen H, van de Vyver M, Ferris WF. Vanadate Impedes Adipogenesis in Mesenchymal Stem Cells Derived from Different Depots within Bone. Front Endocrinol (Lausanne) 2016; 7:108. [PMID: 27536268 PMCID: PMC4971437 DOI: 10.3389/fendo.2016.00108] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/22/2016] [Indexed: 01/01/2023] Open
Abstract
Glucocorticoid-induced osteoporosis (GIO) is associated with an increase in bone marrow adiposity, which skews the differentiation of mesenchymal stem cell (MSC) progenitors away from osteoblastogenesis and toward adipogenesis. We have previously found that vanadate, a non-specific protein tyrosine phosphatase inhibitor, prevents GIO in rats, but it was unclear whether vanadate directly influenced adipogenesis in bone-derived MSCs. For the present study, we investigated the effect of vanadate on adipogenesis in primary rat MSCs derived from bone marrow (bmMSCs) and from the proximal end of the femur (pfMSCs). By passage 3 after isolation, both cell populations expressed the MSC cell surface markers CD90 and CD106, but not the hematopoietic marker CD45. However, although variable, expression of the fibroblast marker CD26 was higher in pfMSCs than in bmMSCs. Differentiation studies using osteogenic and adipogenic induction media (OM and AM, respectively) demonstrated that pfMSCs rapidly accumulated lipid droplets within 1 week of exposure to AM, while bmMSCs isolated from the same femur only formed lipid droplets after 3 weeks of AM treatment. Conversely, pfMSCs exposed to OM produced mineralized extracellular matrix (ECM) after 3 weeks, compared to 1 week for OM-treated bmMSCs. Vanadate (10 μM) added to AM resulted in a significant reduction in AM-induced intracellular lipid accumulation and expression of adipogenic gene markers (PPARγ2, aP2, adipsin) in both pfMSCs and bmMSCs. Pharmacological concentrations of glucocorticoids (1 μM) alone did not induce lipid accumulation in either bmMSCs or pfMSCs, but resulted in significant cell death in pfMSCs. Our findings demonstrate the existence of at least two fundamentally different MSC depots within the femur and highlights the presence of MSCs capable of rapid adipogenesis within the proximal femur, an area prone to osteoporotic fractures. In addition, our results suggest that the increased bone marrow adiposity observed in GIO may not be solely due to direct effect of glucocorticoids on bone-derived MSCs, and that an increase in femur lipid content may also arise from increased adipogenesis in MSCs residing outside of the bone marrow niche.
Collapse
Affiliation(s)
- Frans Alexander Jacobs
- Department of Medicine, Division of Endocrinology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
| | - Hanél Sadie-Van Gijsen
- Department of Medicine, Division of Endocrinology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
| | - Mari van de Vyver
- Department of Medicine, Division of Endocrinology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
| | - William Frank Ferris
- Department of Medicine, Division of Endocrinology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
- *Correspondence: William Frank Ferris,
| |
Collapse
|
129
|
Bosetti M, Sabbatini M, Calarco A, Borrone A, Peluso G, Cannas M. Effect of retinoic acid and vitamin D3 on osteoblast differentiation and activity in aging. J Bone Miner Metab 2016; 34:65-78. [PMID: 25691285 DOI: 10.1007/s00774-014-0642-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 11/24/2014] [Indexed: 02/04/2023]
Abstract
Several studies have evidenced that in aging, osteoblast functional activity is impaired: osteoblast proliferation is slower and matrix deposition is less efficient. Because peroxisome-proliferator-activated receptor γ2 (PPARγ2) and fatty acids are important inhibitory signals in osteoblast development, we have investigated in human primary osteoblasts obtained from patients of different ages, the influence of retinoic acid and calcitriol on enzymes involved in differentiative (PPARγ2, β-catenin, and insulin-like growth factor 1) and metabolic (carnitine palmitoyltransferase 1) intracellular pathways, and on transglutaminase 2, as enzyme fundamental for stabilizing the newly deposited extracellular matrix in bone. Retinoic acid and calcitriol influenced, respectively, proliferation and differentiation of osteoblasts, and an increase in PPARγ2 expression was observed following retinoic acid administration, whereas a decrease was observed following calcitriol administration. Aging widely influenced all parameters analyzed (the proliferation, differentiation, and new matrix deposition are significantly reduced in aged osteoblasts), with the exception of PPARγ2, which we found to be constitutively overexpressed and not modulated by retinoic acid or calcitriol administration. Our findings show the impaired ability of aged osteoblasts to perform adequate functional response and draw attention to the therapeutic approaches for bone healing in elderly patients.
Collapse
Affiliation(s)
- Michela Bosetti
- Pharmacy Science Department, University of Eastern Piedmont, Alessandria, Novara, Vercelli, Italy
| | - Maurizio Sabbatini
- Department of Health Sciences, University of Eastern Piedmont, Alessandria, Novara, Vercelli, Italy.
- Dipartmento Scienze della Salute, Università del Piemonte Orientale "Amedeo Avogadro", via Solaroli 17, 28100, Novara, Italy.
| | - Anna Calarco
- Institute of Protein Biochemistry, CNR, Naples, Italy
| | - Alessia Borrone
- Department of Health Sciences, University of Eastern Piedmont, Alessandria, Novara, Vercelli, Italy
| | | | - Mario Cannas
- Department of Health Sciences, University of Eastern Piedmont, Alessandria, Novara, Vercelli, Italy
| |
Collapse
|
130
|
Sulston RJ, Learman BS, Zhang B, Scheller EL, Parlee SD, Simon BR, Mori H, Bree AJ, Wallace RJ, Krishnan V, MacDougald OA, Cawthorn WP. Increased Circulating Adiponectin in Response to Thiazolidinediones: Investigating the Role of Bone Marrow Adipose Tissue. Front Endocrinol (Lausanne) 2016; 7:128. [PMID: 27708617 PMCID: PMC5030308 DOI: 10.3389/fendo.2016.00128] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/05/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Bone marrow adipose tissue (MAT) contributes to increased circulating adiponectin, an insulin-sensitizing hormone, during caloric restriction (CR), but whether this occurs in other contexts remains unknown. The antidiabetic thiazolidinediones (TZDs) also promote MAT expansion and hyperadiponectinemia, even without increasing adiponectin expression in white adipose tissue (WAT). OBJECTIVES To test the hypothesis that MAT expansion contributes to TZD-associated hyperadiponectinemia, we investigated the effects of rosiglitazone, a prototypical TZD, in wild-type (WT) or Ocn-Wnt10b mice. The latter resist MAT expansion during CR, leading us to postulate that they would also resist this effect of rosiglitazone. DESIGN Male and female WT or Ocn-Wnt10b mice (C57BL/6J) were treated with or without rosiglitazone for 2, 4, or 8 weeks, up to 30 weeks of age. MAT content was assessed by osmium tetroxide staining and adipocyte marker expression. Circulating adiponectin was determined by ELISA. RESULTS In WT mice, rosiglitazone caused hyperadiponectinemia and MAT expansion. Compared to WT mice, Ocn-Wnt10b mice had significantly less MAT in distal tibiae and sometimes in proximal tibiae; however, interpretation was complicated by the leakage of osmium tetroxide from ruptures in some tibiae, highlighting an important technical consideration for osmium-based MAT analysis. Despite decreased MAT in Ocn-Wnt10b mice, circulating adiponectin was generally similar between WT and Ocn-Wnt10b mice; however, in females receiving rosiglitazone for 4 weeks, hyperadiponectinemia was significantly blunted in Ocn-Wnt10b compared to WT mice. Notably, this was also the only group in which tibial adiponectin expression was lower than in WT mice, suggesting a close association between MAT adiponectin production and circulating adiponectin. However, rosiglitazone significantly increased adiponectin protein expression in WAT, suggesting that WAT contributes to hyperadiponectinemia in this context. Finally, rosiglitazone upregulated uncoupling protein 1 in brown adipose tissue (BAT), but this protein was undetectable in tibiae, suggesting that MAT is unlikely to share thermogenic properties of BAT. CONCLUSION TZD-induced hyperadiponectinemia is closely associated with increased adiponectin production in MAT but is not prevented by the partial loss of MAT that occurs in Ocn-Wnt10b mice. Thus, more robust loss-of-MAT models are required for future studies to better establish MAT's elusive functions, both on an endocrine level and beyond.
Collapse
Affiliation(s)
- Richard J. Sulston
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Brian S. Learman
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Bofeng Zhang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Erica L. Scheller
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sebastian D. Parlee
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Becky R. Simon
- Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hiroyuki Mori
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Adam J. Bree
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Venkatesh Krishnan
- Musculoskeletal Research, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Ormond A. MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - William P. Cawthorn
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Musculoskeletal Research, Lilly Research Laboratories, Indianapolis, IN, USA
- *Correspondence: William P. Cawthorn,
| |
Collapse
|
131
|
Weilner S, Keider V, Winter M, Harreither E, Salzer B, Weiss F, Schraml E, Messner P, Pietschmann P, Hildner F, Gabriel C, Redl H, Grillari-Voglauer R, Grillari J. Vesicular Galectin-3 levels decrease with donor age and contribute to the reduced osteo-inductive potential of human plasma derived extracellular vesicles. Aging (Albany NY) 2016; 8:16-33. [PMID: 26752347 PMCID: PMC4761711 DOI: 10.18632/aging.100865] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 11/29/2015] [Indexed: 12/21/2022]
Abstract
Aging results in a decline of physiological functions and in reduced repair capacities, in part due to impaired regenerative power of stem cells, influenced by the systemic environment. In particular osteogenic differentiation capacity (ODC) of mesenchymal stem cells (MSCs) has been shown to decrease with age, thereby contributing to reduced bone formation and an increased fracture risk. Searching for systemic factors that might contribute to this age related decline of regenerative capacity led us to investigate plasma-derived extracellular vesicles (EVs). EVs of the elderly were found to inhibit osteogenesis compared to those of young individuals. By analyzing the differences in the vesicular content Galectin-3 was shown to be reduced in elderly-derived vesicles. While overexpression of Galectin-3 resulted in an enhanced ODC of MSCs, siRNA against Galectin-3 reduced osteogenesis. Modulation of intravesicular Galectin-3 levels correlated with an altered osteo-inductive potential indicating that vesicular Galectin-3 contributes to the biological response of MSCs to EVs. By site-directed mutagenesis we identified a phosphorylation-site on Galectin-3 mediating this effect. Finally, we showed that cell penetrating peptides comprising this phosphorylation-site are sufficient to increase ODC in MSCs. Therefore, we suggest that decrease of Galectin-3 in the plasma of elderly contributes to the age-related loss of ODC.
Collapse
Affiliation(s)
- Sylvia Weilner
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
- Evercyte GmbH, 1190 Vienna, Austria
| | - Verena Keider
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Melanie Winter
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Eva Harreither
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Benjamin Salzer
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Florian Weiss
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Elisabeth Schraml
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Paul Messner
- Department of Nanobiotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
| | - Peter Pietschmann
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Florian Hildner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Christian Gabriel
- Red Cross Blood Transfusion Service of Upper Austria, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Regina Grillari-Voglauer
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
- Evercyte GmbH, 1190 Vienna, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria
- Evercyte GmbH, 1190 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| |
Collapse
|
132
|
Bukowska J, Ziecik AJ, Laguna J, Gawronska-Kozak B, Bodek G. The Importance of the Canonical Wnt Signaling Pathway in the Porcine Endometrial Stromal Stem/Progenitor Cells: Implications for Regeneration. Stem Cells Dev 2015; 24:2873-85. [PMID: 26414529 PMCID: PMC4683563 DOI: 10.1089/scd.2015.0078] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 09/22/2015] [Indexed: 12/12/2022] Open
Abstract
The regenerative ability of the endometrium is strongly associated with the presence of adult stem/progenitor cells. Purposes of the present study were (1) to establish the presence of stem/progenitor cells in porcine endometrial stroma using a clonogenic assay and (2) to investigate whether the canonical Wnt pathway affects the potential of stem/progenitor cells to undergo self-renewal or differentiation. The utility of endometrial stromal clones as a model for stem/progenitor studies was evaluated based on these cells' increased expression of mesenchymal stem cell (MSC) marker genes, including CD29, CD73, CD90, and CD105, compared with primary cultured cells. Small molecules were introduced to activate (BIO) or inhibit (XAV939) the canonical Wnt pathway during stromal clone formation. Cloning efficiency assays revealed that activation of the Wnt/β-catenin pathway promoted formation of more differentiated small clones. Moreover, activation of the Wnt/β-catenin pathway decreased, whereas inhibition of the pathway increased MSC marker expression. Additionally, we confirmed the importance of canonical Wnt pathway stimulation in endometrial stromal cells through observing the appropriate changes in β-catenin cellular localization. These data indicate that modulation of the canonical Wnt pathway effects the process of regeneration in the porcine endometrium during the course of the estrous cycle.
Collapse
Affiliation(s)
- Joanna Bukowska
- In Vitro and Cell Biotechnology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Adam Janusz Ziecik
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Laguna
- In Vitro and Cell Biotechnology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Barbara Gawronska-Kozak
- Department of Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Gabriel Bodek
- In Vitro and Cell Biotechnology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
133
|
Molecular mechanisms of osteoporotic hip fractures in elderly women. Exp Gerontol 2015; 73:49-58. [PMID: 26608808 DOI: 10.1016/j.exger.2015.11.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/28/2015] [Accepted: 11/19/2015] [Indexed: 11/24/2022]
Abstract
A common manifestation of age-related bone loss and resultant osteoporosis are fractures of the hip. Age-related osteoporosis is thought to be determined by a number of intrinsic factors including genetics, hormonal changes, changes in levels of oxidative stress, or an inflammatory status associated with the aging process. The aim of this study was to investigate gene expression and bone architecture in bone samples derived from elderly osteoporotic women with hip fractures (OP) in comparison to bone samples from age matched women with osteoarthritis of the hip (OA). Femoral heads and adjacent neck tissue were collected from 10 women with low-trauma hip fractures (mean age 83±6) and consecutive surgical hip replacement. Ten bone samples from patients undergoing hip replacement due to osteoarthritis (mean age 80±5) served as controls. One half of each bone sample was subjected to gene expression analysis. The second half of each bone sample was analyzed by microcomputed tomography. From each half, samples from four different regions, the central and subcortical region of the femoral head and neck, were analyzed. We could show a significantly decreased expression of the osteoblast related genes RUNX2, Osterix, Sclerostin, WNT10B, and Osteocalcin, a significantly increased ratio of RANKL to Osteoprotegerin, and a significantly increased expression of the enzymes superoxide dismutase 2 (SOD2) and glutathione peroxidase GPX3, and of the inflammatory cytokine IL6 in bone samples from hip fracture patients compared to controls. Major microstructural changes in OP bone were seen in the neck and were characterized by a significant decrease of bone volume, trabecular number, and connectivity density and a significant increase of trabecular separation. In conclusion, our data give evidence for a decreased expression of osteoblast related genes and increased expression of osteoclast related genes. Furthermore, increased expression of SOD2 and GPX3 suggest increased antioxidative activity in bone samples from elderly osteoporotic women with hip fractures.
Collapse
|
134
|
Niedźwiedzki T, Filipowska J. Bone remodeling in the context of cellular and systemic regulation: the role of osteocytes and the nervous system. J Mol Endocrinol 2015; 55:R23-36. [PMID: 26307562 DOI: 10.1530/jme-15-0067] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/24/2015] [Indexed: 12/30/2022]
Abstract
Bone is a dynamic tissue that undergoes constant remodeling. The appropriate course of this process determines development and regeneration of the skeleton. Tight molecular control of bone remodeling is vital for the maintenance of appropriate physiology and microarchitecture of the bone, providing homeostasis, also at the systemic level. The process of remodeling is regulated by a rich innervation of the skeleton, being the source of various growth factors, neurotransmitters, and hormones regulating function of the bone. Although the course of bone remodeling at the cellular level is mainly associated with the activity of osteoclasts and osteoblasts, recently also osteocytes have gained a growing interest as the principal regulators of bone turnover. Osteocytes play a significant role in the regulation of osteogenesis, releasing sclerostin (SOST), an inhibitor of bone formation. The process of bone turnover, especially osteogenesis, is also modulated by extra-skeletal molecules. Proliferation and differentiation of osteoblasts are promoted by the brain-derived serotonin and hypothetically inhibited by its intestinal equivalent. The activity of SOST and serotonin is either directly or indirectly associated with the canonical Wnt/β-catenin signaling pathway, the main regulatory pathway of osteoblasts function. The impairment of bone remodeling may lead to many skeletal diseases, such as high bone mass syndrome or osteoporosis. In this paper, we review the most recent data on the cellular and molecular mechanisms of bone remodeling control, with particular emphasis on the role of osteocytes and the nervous system in this process.
Collapse
Affiliation(s)
- Tadeusz Niedźwiedzki
- Department of Orthopedics and PhysiotherapyCollegium Medicum, Jagiellonian University, Cracow, PolandDepartment of Cell Biology and ImagingInstitute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Cracow, Poland
| | - Joanna Filipowska
- Department of Orthopedics and PhysiotherapyCollegium Medicum, Jagiellonian University, Cracow, PolandDepartment of Cell Biology and ImagingInstitute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Cracow, Poland
| |
Collapse
|
135
|
Lu J, Duan Y, Zhang M, Wu M, Wang Y. Expression of Wnt3a, Wnt10b, β-catenin and DKK1 in periodontium during orthodontic tooth movement in rats. Acta Odontol Scand 2015; 74:217-23. [PMID: 26414930 DOI: 10.3109/00016357.2015.1090011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To investigate the expression of Wnt3a, Wnt10b, β-catenin and DKK1 in the periodontal ligament (PDL) during orthodontic tooth movement (OTM) in rats. MATERIALS AND METHODS Nickel-titanium closed-coil springs were used to deliver an initial 50 g mesial force to the left maxillary first molars in 30 rats. The force was kept constant for 1, 3, 5, 7, 10 and 14 days until the animals were sacrificed. The right maxillary molars without force application served as control. Paraffin-embedded sections of the upper jaws were prepared for histological and immunohistochemical analyses to detect Wnt3a, Wnt10b, β-catenin and DKK1 expression in PDL. RESULTS Wnt3a, Wnt10b, β-catenin and DKK1 were expressed on both the ipsilateral and contralateral sides of PDL in each group. After the application of orthodontic force, the expression of β-catenin and DKK1 was initially increased and then decreased on both sides, with maximal levels of expression at day 7 and day 10, respectively. On the compression side, Wnt3a and Wnt10b levels started to increase at day 5, while on the tension side, these two molecules began to increase at day 1. Furthermore, the expression levels of Wnt3a, Wnt10b, and β-catenin were much stronger on the tension side than on the compression side at any of the observation points, while DKK1 level was much higher on the compression side. CONCLUSION Wnt3a, Wnt10b, β-catenin and DKK1 expression may be related to the periodontal tissue remodeling following the application of an orthodontic force in rats. These observations suggest that the Wnt/β-catenin signaling pathway may play a crucial role in periodontal tissue remodeling during OTM.
Collapse
Affiliation(s)
- Juan Lu
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Yingying Duan
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Miaomiao Zhang
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Mingming Wu
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| | - Ying Wang
- a Department of Orthodontics, School of Stomatology , Harbin Medical University , Harbin , 150001 , PR China
| |
Collapse
|
136
|
Single-pulsed electromagnetic field therapy increases osteogenic differentiation through Wnt signaling pathway and sclerostin downregulation. Bioelectromagnetics 2015; 36:494-505. [DOI: 10.1002/bem.21933] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/15/2015] [Indexed: 01/20/2023]
|
137
|
Zhang J, Motyl KJ, Irwin R, MacDougald OA, Britton RA, McCabe LR. Loss of Bone and Wnt10b Expression in Male Type 1 Diabetic Mice Is Blocked by the Probiotic Lactobacillus reuteri. Endocrinology 2015; 156:3169-82. [PMID: 26135835 PMCID: PMC4541610 DOI: 10.1210/en.2015-1308] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes (T1D)-induced osteoporosis is characterized by a predominant suppression of osteoblast number and activity, as well as increased bone marrow adiposity but no change in osteoclast activity. The fundamental mechanisms and alternative anabolic treatments (with few side effects) for T1D bone loss remain undetermined. Recent studies by our laboratory and others indicate that probiotics can benefit bone health. Here, we demonstrate that Lactobacillus reuteri, a probiotic with anti-inflammatory and bone health properties, prevents T1D-induced bone loss and marrow adiposity in mice. We further found that L. reuteri treatment prevented the suppression of Wnt10b in T1D bone. Consistent with a role for attenuated bone Wnt10b expression in T1D osteoporosis, we observed that bone-specific Wnt10b transgenic mice are protected from T1D bone loss. To examine the mechanisms of this protection, we focused on TNF-α, a cytokine up-regulated in T1D that causes suppression of osteoblast Wnt10b expression in vitro. Addition of L. reuteri prevented TNF-α-mediated suppression of Wnt10b and osteoblast maturation markers. Taken together, our findings reveal a mechanism by which T1D causes bone loss and open new avenues for use of probiotics to benefit the bone.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Physiology (J.Z., K.J.M., R.I., L.R.M.), Department of Molecular and Integrative Physiology (O.A.M.), Department of Microbiology and Molecular Genetics (R.A.B.), Department of Radiology (L.R.M.), and Biomedical Imaging Research Center (L.R.M.), Michigan State University, East Lansing, Michigan 48824
| | - Katherine J Motyl
- Department of Physiology (J.Z., K.J.M., R.I., L.R.M.), Department of Molecular and Integrative Physiology (O.A.M.), Department of Microbiology and Molecular Genetics (R.A.B.), Department of Radiology (L.R.M.), and Biomedical Imaging Research Center (L.R.M.), Michigan State University, East Lansing, Michigan 48824
| | - Regina Irwin
- Department of Physiology (J.Z., K.J.M., R.I., L.R.M.), Department of Molecular and Integrative Physiology (O.A.M.), Department of Microbiology and Molecular Genetics (R.A.B.), Department of Radiology (L.R.M.), and Biomedical Imaging Research Center (L.R.M.), Michigan State University, East Lansing, Michigan 48824
| | - Ormond A MacDougald
- Department of Physiology (J.Z., K.J.M., R.I., L.R.M.), Department of Molecular and Integrative Physiology (O.A.M.), Department of Microbiology and Molecular Genetics (R.A.B.), Department of Radiology (L.R.M.), and Biomedical Imaging Research Center (L.R.M.), Michigan State University, East Lansing, Michigan 48824
| | - Robert A Britton
- Department of Physiology (J.Z., K.J.M., R.I., L.R.M.), Department of Molecular and Integrative Physiology (O.A.M.), Department of Microbiology and Molecular Genetics (R.A.B.), Department of Radiology (L.R.M.), and Biomedical Imaging Research Center (L.R.M.), Michigan State University, East Lansing, Michigan 48824
| | - Laura R McCabe
- Department of Physiology (J.Z., K.J.M., R.I., L.R.M.), Department of Molecular and Integrative Physiology (O.A.M.), Department of Microbiology and Molecular Genetics (R.A.B.), Department of Radiology (L.R.M.), and Biomedical Imaging Research Center (L.R.M.), Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
138
|
Zhong ZA, Zahatnansky J, Snider J, Van Wieren E, Diegel CR, Williams BO. Wntless spatially regulates bone development through β-catenin-dependent and independent mechanisms. Dev Dyn 2015; 244:1347-55. [PMID: 26249818 DOI: 10.1002/dvdy.24316] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 06/30/2015] [Accepted: 07/06/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Canonical and noncanonical Wnt signaling pathways both play pivotal roles in bone development. Wntless/GPR177 is a chaperone protein that is required for secretion of all Wnt ligands. We previously showed that deletion of Wntless within mature osteoblasts severely impaired postnatal bone homeostasis. RESULTS In this study, we systemically evaluated how deletion of Wntless in different stages of osteochondral differentiation affected embryonic bone development, by crossing Wntless (Wls)-flox/flox mice with strains expressing cre recombinase behind the following promoters: Osteocalcin, Collagen 2a1, or Dermo1. Ex vivo µCT and whole-mount skeletal staining were performed to examine skeletal mineralization. Histology and immunohistochemistry were used to evaluate cellular differentiation and alterations in Wnt signaling. In this work, we found that Wntless regulated chondrogenesis and osteogenesis through both canonical and noncanonical Wnt signaling. CONCLUSIONS These findings provide more insight into the requirements of different Wnt-secretion cell types critical for skeletal development.
Collapse
Affiliation(s)
- Zhendong A Zhong
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Juraj Zahatnansky
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - John Snider
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Emily Van Wieren
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Cassandra R Diegel
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Bart O Williams
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| |
Collapse
|
139
|
Misu M, Ouji Y, Kawai N, Nishimura F, Nakamura-Uchiyama F, Yoshikawa M. Effects of Wnt-10b on proliferation and differentiation of murine melanoma cells. Biochem Biophys Res Commun 2015; 463:618-23. [PMID: 26056007 DOI: 10.1016/j.bbrc.2015.05.110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 05/29/2015] [Indexed: 12/27/2022]
Abstract
In spite of the strong expression of Wnt-10b in melanomas, its role in melanoma cells has not been elucidated. In the present study, the biological effects of Wnt-10b on murine B16F10 (B16) melanoma cells were investigated using conditioned medium from Wnt-10b-producing COS cells (Wnt-CM). After 2 days of culture in the presence of Wnt-CM, proliferation of B16 melanoma cells was inhibited, whereas tyrosinase activity was increased. An in vitro wound healing assay demonstrated that migration of melanoma cells to the wound area was inhibited with the addition of Wnt-CM. Furthermore, evaluation of cellular senescence revealed prominent induction of SA-β-gal-positive senescent cells in cultures with Wnt-CM. Finally, the growth of B16 melanoma cell aggregates in collagen 3D-gel cultures was markedly suppressed in the presence of Wnt-CM. These results suggest that Wnt-10b represses tumor cell properties, such as proliferation and migration of B16 melanoma cells, driving them toward a more differentiated state along a melanocyte lineage.
Collapse
Affiliation(s)
- Masayasu Misu
- Department of Pathogen, Infection and Immunity, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Yukiteru Ouji
- Department of Pathogen, Infection and Immunity, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan.
| | - Norikazu Kawai
- Department of Pathogen, Infection and Immunity, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Fumihiko Nishimura
- Department of Neurosurgery, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Fukumi Nakamura-Uchiyama
- Department of Pathogen, Infection and Immunity, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Masahide Yoshikawa
- Department of Pathogen, Infection and Immunity, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan.
| |
Collapse
|
140
|
Zhang F, Zhang Z, Sun D, Dong S, Xu J, Dai F. EphB4 Promotes Osteogenesis of CTLA4-Modified Bone Marrow-Derived Mesenchymal Stem Cells Through Cross Talk with Wnt Pathway in Xenotransplantation. Tissue Eng Part A 2015; 21:2404-16. [PMID: 26132739 DOI: 10.1089/ten.tea.2015.0012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cytotoxic T lymphocyte-associated antigen 4-Ig (CTLA4-Ig)-modified bone marrow-derived mesenchymal stem cells (MSCs-CTLA4) have excellent osteogenic function in xenografts, but their mechanism of action remains to be elucidated. As bidirectional signaling between erythropoietin-producing hepatocyte receptors B4 (EphB4) and ephrinB2 is vital for bone remodeling, this study aimed to fully characterize the role of MSCs-CTLA4 in promoting bone regeneration in xenotransplantation through EphB4/ephrinB2 and their cross talk with the Wnt/beta-catenin pathway. METHODS MSCs-CTLA4 were investigated for their osteogenic capacity through xenotransplantation in vivo. MSCs-CTLA4 were treated with ephrinB2-FC or FC under conditions of osteogenic induction and cultured with or without immune activation conditions established by phytohemagglutinin and peripheral blood mononuclear cells in vitro. Osteogenesis markers and the Wnt pathway-related molecules such as EphB4, runt-related transcription factor 2 (Runx2), collagen 1 (COL1), osteocalcin (OCN), alkaline phosphatase (ALP), calcium nodus, β-catenin, phospho-glycogen synthase kinase 3-beta (p-GSK-3β)-Ser9, and glycogen synthase kinase 3-beta (GSK-3β) were detected. RESULTS MSCs-CTLA4-based xenografts show better osteogenic capacity compared with MSC-based xenografts. EphB4 expression was reduced in MSCs compared with MSCs-CTLA4 under immune activation conditions. In ephrinB2-FC-treated cells, levels of osteogenesis markers were increased compared with FC-treated cells. The activity of GSK-3 was inhibited and the expression of β-catenin in MSCs was increased by ephrinB2-FC treatment. CONCLUSIONS CTLA4 modification maintains EphB4 expression in MSCs under immune activation conditions, and EphB4 cross talk with the Wnt pathway promotes osteogenic differentiation of MSCs-CTLA4.
Collapse
Affiliation(s)
- Fei Zhang
- 1 National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University , Chongqing, People's Republic of China .,2 Department of Orthopaedics, Southwest Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Zehua Zhang
- 1 National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University , Chongqing, People's Republic of China .,2 Department of Orthopaedics, Southwest Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Dong Sun
- 1 National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University , Chongqing, People's Republic of China .,2 Department of Orthopaedics, Southwest Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Shiwu Dong
- 1 National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University , Chongqing, People's Republic of China .,3 Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University , Chongqing, People's Republic of China
| | - Jianzhong Xu
- 1 National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University , Chongqing, People's Republic of China .,2 Department of Orthopaedics, Southwest Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Fei Dai
- 1 National & Regional United Engineering Laboratory of Tissue Engineering, Third Military Medical University , Chongqing, People's Republic of China .,2 Department of Orthopaedics, Southwest Hospital, Third Military Medical University , Chongqing, People's Republic of China
| |
Collapse
|
141
|
Coe LM, Tekalur SA, Shu Y, Baumann MJ, McCabe LR. Bisphosphonate treatment of type I diabetic mice prevents early bone loss but accentuates suppression of bone formation. J Cell Physiol 2015; 230:1944-53. [PMID: 25641511 DOI: 10.1002/jcp.24929] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
Type I (T1) diabetes is an autoimmune and metabolic disease associated with bone loss. Previous studies demonstrate that T1-diabetes decreases osteoblast activity and viability. Bisphosphonate therapy, commonly used to treat osteoporosis, is demonstrated to inhibit osteoclast activity as well as osteoblast apoptosis. Therefore, we examined the effect of weekly alendronate treatments on T1-diabetes induced osteoblast apoptosis and bone loss. Bone TUNEL assays identified that alendronate therapy prevents the diabetes-induced osteoblast death observed during early stages of diabetes development. Consistent with this, alendronate treatment for 40 days was able to prevent diabetes-induced trabecular bone loss. Alendronate was also able to reduce marrow adiposity in both control diabetic mice compared to untreated mice. Mechanical testing indicated that 40 days of alendronate treatment increased bone stiffness but decreased the work required for fracture in T1-diabetic and alendronate treated mice. Of concern at this later time point, bone formation rate and osteoblast markers, which were already decreased in diabetic mice, were further suppressed in alendronate-treated diabetic mice. Taken together, our results suggest that short-term alendronate treatment can prevent T1-diabetes-induced bone loss in mice, possibly in part by inhibiting diabetes onset associated osteoblast death, while longer treatment enhanced bone density but at the cost of further suppressing bone formation in diabetic mice.
Collapse
Affiliation(s)
- Lindsay M Coe
- Department of Physiology, Biomedical Imaging Research Center, Michigan State University, East Lansing, Michigan
| | | | | | | | | |
Collapse
|
142
|
Qin Z, Fang Z, Zhao L, Chen J, Li Y, Liu G. High dose of TNF-α suppressed osteogenic differentiation of human dental pulp stem cells by activating the Wnt/β-catenin signaling. J Mol Histol 2015; 46:409-20. [PMID: 26115593 DOI: 10.1007/s10735-015-9630-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/23/2015] [Indexed: 12/31/2022]
Abstract
Dental pulp stem cells (DPSCs) were a clonogenic, highly proliferative cells capable of self-renewal and multi-lineage differentiation including chondrocytes, adipocytes, neural cells and osteoblasts, which make it an attractive choice for bone regeneration and repair of craniofacial defects. Recent studies showed that tumor necrosis factor α (TNF-α) may affect osteoclastogenesis and bone formation. However, the effect and mechanism of TNF-α on DPSCs is not clear. In this study, we found that low dose TNF-α promoted mineralization and high dose TNF-α suppressed osteogenic differentiation of DPSCs. Levels of ALP, Osteopontin, Osteocalcin, Osterix and Runx2 were up-regulated in DPSCs treated with TNF-α at low concentration, while down-regulated in DPSCs treated with TNF-α at high concentration. Blockade of Wnt/β-catenin signaling reversed the inhibitory effect observed on osteogenic differentiation of DPSCs treated with TNF-α at high concentration. In addition, we did not detect any proliferative effect of TNF-α on DPSCs by cell cycle and cell counts analysis. In summary, our data suggested that high concentration TNF-α suppressed mineralization and mineralization-related gene expressions through the Wnt/β-catenin signaling in DPSCs. Our findings may provide a foundation for autologous transplantation of DPSCs.
Collapse
Affiliation(s)
- Zhenjie Qin
- Department of Stomatology, Zoucheng People's Hospital, Zoucheng, 273500, Shandong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
143
|
Abstract
WNTs are extracellular proteins that activate different cell surface receptors linked to canonical and noncanonical WNT signalling pathways. The Wnt genes were originally discovered as important for embryonic development of fruit flies and malignant transformation of mouse mammary cancers. More recently, WNTs have been implicated in a wide spectrum of biological phenomena and diseases. During the last decade, several lines of clinical and preclinical evidence have indicated that WNT signalling is critical for trabecular and cortical bone mass, and this pathway is currently an attractive target for drug development. Based on detailed knowledge of the different WNT signalling pathways, it appears that it might be possible to develop drugs that specifically target cortical and trabecular bone. Neutralization of a bone-specific WNT inhibitor is now being evaluated as a promising anabolic treatment for patients with osteoporosis. Here, we provide the historical background to the discoveries of WNTs, describe the different WNT signalling pathways and summarize the current understanding of how these proteins regulate bone mass by affecting bone formation and resorption.
Collapse
Affiliation(s)
- U H Lerner
- Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - C Ohlsson
- Centre for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
144
|
Jules J, Yang S, Chen W, Li YP. Role of Regulators of G Protein Signaling Proteins in Bone Physiology and Pathophysiology. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 133:47-75. [PMID: 26123302 DOI: 10.1016/bs.pmbts.2015.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Regulators of G protein signaling (RGS) proteins enhance the intrinsic GTPase activity of α subunits of the heterotrimeric G protein complex of G protein-coupled receptors (GPCRs) and thereby inactivate signal transduction initiated by GPCRs. The RGS family consists of nearly 37 members with a conserved RGS homology domain which is critical for their GTPase accelerating activity. RGS proteins are expressed in most tissues, including heart, lung, brain, kidney, and bone and play essential roles in many physiological and pathological processes. In skeletal development and bone homeostasis as well as in many bone disorders, RGS proteins control the functions of various GPCRs, including the parathyroid hormone receptor type 1 and calcium-sensing receptor and also regulate various critical signaling pathways, such as Wnt and calcium oscillations. This chapter will discuss the current findings on the roles of RGS proteins in regulating signaling of key GPCRs in skeletal development and bone homeostasis. We also will examine the current updates of RGS proteins' regulation of calcium oscillations in bone physiology and highlight the roles of RGS proteins in selected bone pathological disorders. Despite the recent advances in bone and mineral research, RGS proteins remain understudied in the skeletal system. Further understanding of the roles of RGS proteins in bone should not only provide great insights into the molecular basis of various bone diseases but also generate great therapeutic drug targets for many bone diseases.
Collapse
Affiliation(s)
- Joel Jules
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shuying Yang
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, New York, USA; Developmental Genomics Group, New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Wei Chen
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yi-Ping Li
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
145
|
Jing W, Smith AA, Liu B, Li J, Hunter DJ, Dhamdhere G, Salmon B, Jiang J, Cheng D, Johnson CA, Chen S, Lee K, Singh G, Helms JA. Reengineering autologous bone grafts with the stem cell activator WNT3A. Biomaterials 2015; 47:29-40. [DOI: 10.1016/j.biomaterials.2014.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/29/2014] [Accepted: 12/16/2014] [Indexed: 01/12/2023]
|
146
|
Choudhary S, Canalis E, Estus T, Adams D, Pilbeam C. Cyclooxygenase-2 suppresses the anabolic response to PTH infusion in mice. PLoS One 2015; 10:e0120164. [PMID: 25781979 PMCID: PMC4363701 DOI: 10.1371/journal.pone.0120164] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 02/03/2015] [Indexed: 11/29/2022] Open
Abstract
We previously reported that the ability of continuously elevated PTH to stimulate osteoblastic differentiation in bone marrow stromal cell cultures was abrogated by an osteoclastic factor secreted in response to cyclooxygenase-2 (Cox2)-produced prostaglandin E2. We now examine the impact of Cox2 (Ptgs2) knockout (KO) on the anabolic response to continuously elevated PTH in vivo. PTH (40 μg/kg/d) or vehicle was infused for 12 or 21 days in 3-mo-old male wild type (WT) and KO mice in the outbred CD-1 background. Changes in bone phenotype were assessed by bone mineral density (BMD), μCT and histomorphometry. PTH infusion for both 12 and 21 days increased femoral BMD in Cox2 KO mice and decreased BMD in WT mice. Femoral and vertebral trabecular bone volume fractions were increased in KO mice, but not in WT mice, by PTH infusion. In the femoral diaphysis, PTH infusion increased cortical area in Cox2 KO, but not WT, femurs. PTH infusion markedly increased trabecular bone formation rate in the femur, serum markers of bone formation, and expression of bone formation-related genes, growth factors, and Wnt target genes in KO mice relative to WT mice, and decreased gene expression of Wnt antagonists only in KO mice. In contrast to the differential effects of PTH on anabolic factors in WT and KO mice, PTH infusion increased serum markers of resorption, expression of resorption-related genes, and the percent bone surface covered by osteoclasts similarly in both WT and KO mice. We conclude that Cox2 inhibits the anabolic, but not the catabolic, effects of continuous PTH. These data suggest that the bone loss with continuously infused PTH in mice is due largely to suppression of bone formation and that this suppression is mediated by Cox2.
Collapse
Affiliation(s)
- Shilpa Choudhary
- New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Ernesto Canalis
- New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Thomas Estus
- New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Douglas Adams
- New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Carol Pilbeam
- New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
147
|
Kook SH, Heo JS, Lee JC. Crucial roles of canonical Runx2-dependent pathway on Wnt1-induced osteoblastic differentiation of human periodontal ligament fibroblasts. Mol Cell Biochem 2015; 402:213-23. [DOI: 10.1007/s11010-015-2329-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/16/2015] [Indexed: 10/24/2022]
|
148
|
Du HM, Wang LY, Zheng XH, Tang W, Liu L, Jing W, Lin YF, Tian WD, Long J. The Role of the Wnt Signaling Pathway in the Osteogenic Differentiation of Human Adipose-derived Stem Cells under Mechanical Stimulation. J HARD TISSUE BIOL 2015; 24:169-180. [DOI: 10.2485/jhtb.24.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Hong-ming Du
- The State Key Laboratory of Oral Diseases, Sichuan University
| | - Li-ya Wang
- Department of Stomatology, The First Affiliated Hospital of Soochow University
| | - Xiao-hui Zheng
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University
| | - Wei Tang
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University
| | - Lei Liu
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University
| | - Wei Jing
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University
| | - Yun-feng Lin
- The State Key Laboratory of Oral Diseases, Sichuan University
| | - Wei-dong Tian
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University
| | - Jie Long
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University
- The State Key Laboratory of Oral Diseases, Sichuan University
| |
Collapse
|
149
|
Wnts produced by Osterix-expressing osteolineage cells regulate their proliferation and differentiation. Proc Natl Acad Sci U S A 2014; 111:E5262-71. [PMID: 25422448 DOI: 10.1073/pnas.1420463111] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Wnt signaling is a critical regulator of bone development, but the identity and role of the Wnt-producing cells are still unclear. We addressed these questions through in situ hybridization, lineage tracing, and genetic experiments. First, we surveyed the expression of all 19 Wnt genes and Wnt target gene Axin2 in the neonatal mouse bone by in situ hybridization, and demonstrated--to our knowledge for the first time--that Osterix-expressing cells coexpress Wnt and Axin2. To track the behavior and cell fate of Axin2-expressing osteolineage cells, we performed lineage tracing and showed that they sustain bone formation over the long term. Finally, to examine the role of Wnts produced by Osterix-expressing cells, we inhibited Wnt secretion in vivo, and observed inappropriate differentiation, impaired proliferation, and diminished Wnt signaling response. Therefore, Osterix-expressing cells produce their own Wnts that in turn induce Wnt signaling response, thereby regulating their proliferation and differentiation.
Collapse
|
150
|
Lentivirus-mediated Wnt10b overexpression enhances fracture healing in a rat atrophic non-union model. Biotechnol Lett 2014; 37:733-9. [DOI: 10.1007/s10529-014-1703-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
|