1
|
Thakore P, Karki S, Hrdlicka HC, Garcia-Munoz J, Pereira RC, Delany AM. Decreasing miR-433-3p Activity in the Osteoblast Lineage Blunts Glucocorticoid-mediated Bone Loss. Endocrinology 2025; 166:bqaf008. [PMID: 39820728 PMCID: PMC11791524 DOI: 10.1210/endocr/bqaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
Glucocorticoid excess causes bone loss due to decreased bone formation and increased bone resorption; miR-433-3p is a microRNA (miRNA) that negatively regulates bone formation in male mice by targeting Runx2 as well as RNAs involved in Wnt, protein kinase A, and endogenous glucocorticoid signaling. To examine the impact of miR-433-3p on glucocorticoid-mediated bone loss, transgenic mice expressing a miR-433-3p tough decoy inhibitor in the osteoblast lineage were administered prednisolone via slow-release pellets. Bone loss was greater in control mice treated with prednisolone compared with miR-433-3p tough decoy mice due to higher osteoclast activity in the controls. In whole femurs, Rankl was significantly higher in prednisolone-treated controls compared with miR-433-3p tough decoy mice. Surprisingly, negative regulators of Wnt signaling Sost and Dkk1 were higher in miR-433-3p tough decoy mice and were unaffected by prednisolone. Luciferase- 3'-untranslated region reporter assays demonstrated that Sost is a novel miR-433-3p target, whereas Dkk1 is a previously validated miR-433-3p target. miR-433-3p levels are lower in matrix-synthesizing osteoblasts than in more osteocytic cells; thus the impact of miR-433-3p on the osteoblast lineage may be dependent on cell context: it is a negative regulator in matrix-depositing osteoblasts by targeting RNAs important for differentiation and function but a positive regulator in osteocytes, due to its ability to target prominently expressed negative regulators of Wnt signaling, Sost and Dkk1. The mechanisms by which miR-433-3p indirectly regulates glucocorticoid-mediated osteoclastogenesis remain unknown. However, we speculate that this regulation may be mediated by miR-433-3p activity in osteocytes, which play an important role in controlling osteoclastogenesis.
Collapse
Affiliation(s)
- Prachi Thakore
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Sangita Karki
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - John Garcia-Munoz
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Renata C Pereira
- Division of Pediatric Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
2
|
Yee CS, Meliadis C, Kaya S, Chang W, Alliston T. The osteocytic actions of glucocorticoids on bone mass, mechanical properties, or perilacunar remodeling outcomes are not rescued by PTH(1-34). Front Endocrinol (Lausanne) 2024; 15:1342938. [PMID: 39092287 PMCID: PMC11291448 DOI: 10.3389/fendo.2024.1342938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 06/26/2024] [Indexed: 08/04/2024] Open
Abstract
Glucocorticoids (GC) and parathyroid hormone (PTH) are widely used therapeutic endocrine hormones where their effects on bone and joint arise from actions on multiple skeletal cell types. In osteocytes, GC and PTH exert opposing effects on perilacunar canalicular remodeling (PLR). Suppressed PLR can impair bone quality and joint homeostasis, including in GC-induced osteonecrosis. However, combined effects of GC and PTH on PLR are unknown. Given the untapped potential to target osteocytes to improve skeletal health, this study sought to test the feasibility of therapeutically mitigating PLR suppression. Focusing on subchondral bone and joint homeostasis, we hypothesize that PTH(1-34), a PLR agonist, could rescue GC-suppressed PLR. The skeletal effects of GC and PTH(1-34), alone or combined, were examined in male and female mice by micro-computed tomography, mechanical testing, histology, and gene expression analysis. For each outcome, females were more responsive to GC and PTH(1-34) than males. GC and PTH(1-34) exerted regional differences, with GC increasing trabecular bone volume but reducing cortical bone thickness, stiffness, and ultimate force. Despite PTH(1-34)'s anabolic effects on trabecular bone, it did not rescue GC's catabolic effects on cortical bone. Likewise, cartilage integrity and subchondral bone apoptosis, tartrate-resistant acid phosphatase (TRAP) activity, and osteocyte lacunocanalicular networks showed no evidence that PTH(1-34) could offset GC-dependent effects. Rather, GC and PTH(1-34) each increased cortical bone gene expression implicated in bone resorption by osteoclasts and osteocytes, including Acp5, Mmp13, Atp6v0d2, Ctsk, differences maintained when GC and PTH(1-34) were combined. Since PTH(1-34) is insufficient to rescue GC's effects on young female mouse bone, future studies are needed to determine if osteocyte PLR suppression, due to GC, aging, or other factors, can be offset by a PLR agonist.
Collapse
Affiliation(s)
- Cristal S. Yee
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Christoforos Meliadis
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Wenhan Chang
- Endocrine Research Unit, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, CA, United States
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
3
|
Li X, Liang T, Dai B, Chang L, Zhang Y, Hu S, Guo J, Xu S, Zheng L, Yao H, Lian H, Nie Y, Li Y, He X, Yao Z, Tong W, Wang X, Chow DHK, Xu J, Qin L. Excess glucocorticoids inhibit murine bone turnover via modulating the immunometabolism of the skeletal microenvironment. J Clin Invest 2024; 134:e166795. [PMID: 38512413 DOI: 10.1172/jci166795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
Elevated bone resorption and diminished bone formation have been recognized as the primary features of glucocorticoid-associated skeletal disorders. However, the direct effects of excess glucocorticoids on bone turnover remain unclear. Here, we explored the outcomes of exogenous glucocorticoid treatment on bone loss and delayed fracture healing in mice and found that reduced bone turnover was a dominant feature, resulting in a net loss of bone mass. The primary effect of glucocorticoids on osteogenic differentiation was not inhibitory; instead, they cooperated with macrophages to facilitate osteogenesis. Impaired local nutrient status - notably, obstructed fatty acid transportation - was a key factor contributing to glucocorticoid-induced impairment of bone turnover in vivo. Furthermore, fatty acid oxidation in macrophages fueled the ability of glucocorticoid-liganded receptors to enter the nucleus and then promoted the expression of BMP2, a key cytokine that facilitates osteogenesis. Metabolic reprogramming by localized fatty acid delivery partly rescued glucocorticoid-induced pathology by restoring a healthier immune-metabolic milieu. These data provide insights into the multifactorial metabolic mechanisms by which glucocorticoids generate skeletal disorders, thus suggesting possible therapeutic avenues.
Collapse
Affiliation(s)
- Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Tongzhou Liang
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Yuan Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shiwen Hu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Shunxiang Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Hong Lian
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, and
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Xuan He
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Zhi Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Xinluan Wang
- Centre for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| |
Collapse
|
4
|
Chargo NJ, Neugebauer K, Guzior DV, Quinn RA, Parameswaran N, McCabe LR. Glucocorticoid-induced osteoporosis is prevented by dietary prune in female mice. Front Cell Dev Biol 2024; 11:1324649. [PMID: 38375074 PMCID: PMC10875082 DOI: 10.3389/fcell.2023.1324649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 02/21/2024] Open
Abstract
Glucocorticoid-induced osteoporosis (GIO) is a significant side effect of prolonged glucocorticoid (GC) treatment. Chronic GC treatment also leads to trabecular bone loss and gut microbiota dysbiosis in mice. The gut dysbiosis is mechanistically linked to GIO, which indicates that the microbiota can be targeted to prevent GIO. Prunes, a dried fruit and prebiotic, have emerged in the literature as an effective treatment for sex-steroid deficiency induced osteoporosis (primary osteoporosis). Prunes also significantly alter the composition of the gut microbiota in both rodent models and human studies. Therefore, we tested if dietary prune (DP) supplementation could prevent GC-induced bone loss and affect microbiota composition in an established model of GIO. Sixteen-week-old, skeletally mature, female C57BL/6J mice were treated with a subcutaneous 5 mg placebo or prednisolone pellet for 8 weeks and fed an AIN-93M control diet or a diet modified to include 5, 15, or 25% (w/w) dried California prune powder. As expected, GC treated mice developed significant trabecular bone loss in the distal femur. More importantly, as little as 5% DP supplementation effectively prevented trabecular bone loss. Further, dose dependent increases in trabecular bone volume fraction were observed in GC + 15% and GC + 25% DP mice. Amazingly, in the placebo (non-GC treated) groups, 25% DP supplementation caused a ∼3-fold increase in distal femur trabecular bone volume fraction; this sizable bone response has not been previously observed in healthy mice with gut targeted natural treatments. Along with the striking effect on bone health, GC treatment and 25% DP supplementation led to drastic shifts in gut microbiota composition and several specific changes are strongly associated with bone health. Taken together, these results are the first to demonstrate that DP supplementation effectively prevents the negative effects of prolonged GC therapy on trabecular bone health and strongly associates with shifts in the composition of the gut microbiota.
Collapse
Affiliation(s)
- Nicholas J. Chargo
- Department of Physiology, Michigan State University, East Lansing, MI, United States
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| | - Kerri Neugebauer
- Department of Plant Soil and Microbiology, Michigan State University, East Lansing, MI, United States
| | - Douglas V. Guzior
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Robert A. Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, MI, United States
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Laura R. McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, United States
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
5
|
Krüger BT, Steppe L, Vettorazzi S, Haffner-Luntzer M, Lee S, Dorn AK, Ignatius A, Tuckermann J, Ahmad M. Inhibition of Cdk5 Ameliorates Skeletal Bone Loss in Glucocorticoid-Treated Mice. Biomedicines 2022; 10:404. [PMID: 35203613 PMCID: PMC8962259 DOI: 10.3390/biomedicines10020404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids (GCs) are widely used to treat inflammatory diseases. However, their long-term use leads to glucocorticoid-induced osteoporosis, increasing morbidity and mortality. Both anabolic and anti-resorptive drugs are used to counteract GC-induced bone loss, however, they are expensive and/or have major side effects. Therefore, identifying new targets for cost-effective, small-molecule inhibitors is essential. We recently identified cyclin-dependent kinase 5 (Cdk5) as a suppressor of osteoblast differentiation and showed that its inhibition with roscovitine promoted osteoblastogenesis, thus improving the skeletal bone mass and fracture healing. Here, we assessed whether Cdk5 knockdown or inhibition could also reverse the GC-mediated suppression of osteoblast differentiation, bone loss, and fracture healing. We first demonstrated that Cdk5 silencing abolished the dexamethasone (Dex)-induced downregulation of alkaline phosphatase (Alp) activity, osteoblast-specific marker gene expression (Runx2, Sp7, Alpl, and Bglap), and mineralization. Similarly, Cdk5 inhibition rescued Dex-induced suppression of Alp activity. We further demonstrated that Cdk5 inhibition reversed prednisolone (Pred)-induced bone loss in mice, due to reduced osteoclastogenesis rather than improved osteoblastogenesis. Moreover, we revealed that Cdk5 inhibition failed to improve Pred-mediated impaired fracture healing. Taken together, we demonstrated that Cdk5 inhibition with roscovitine ameliorated GC-mediated bone loss but did not reverse GC-induced compromised fracture healing in mice.
Collapse
Affiliation(s)
- Benjamin Thilo Krüger
- Institute of Orthopedic Research and Biomechanics, Ulm University, Helmholtzstrasse 14, 89081 Ulm, Germany; (B.T.K.); (L.S.); (M.H.-L.)
| | - Lena Steppe
- Institute of Orthopedic Research and Biomechanics, Ulm University, Helmholtzstrasse 14, 89081 Ulm, Germany; (B.T.K.); (L.S.); (M.H.-L.)
| | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Helmholtzstrasse 8/1, 89081 Ulm, Germany; (S.V.); (S.L.); (A.-K.D.)
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University, Helmholtzstrasse 14, 89081 Ulm, Germany; (B.T.K.); (L.S.); (M.H.-L.)
| | - Sooyeon Lee
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Helmholtzstrasse 8/1, 89081 Ulm, Germany; (S.V.); (S.L.); (A.-K.D.)
| | - Ann-Kristin Dorn
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Helmholtzstrasse 8/1, 89081 Ulm, Germany; (S.V.); (S.L.); (A.-K.D.)
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University, Helmholtzstrasse 14, 89081 Ulm, Germany; (B.T.K.); (L.S.); (M.H.-L.)
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Helmholtzstrasse 8/1, 89081 Ulm, Germany; (S.V.); (S.L.); (A.-K.D.)
| | - Mubashir Ahmad
- Institute of Orthopedic Research and Biomechanics, Ulm University, Helmholtzstrasse 14, 89081 Ulm, Germany; (B.T.K.); (L.S.); (M.H.-L.)
- Institute of Comparative Molecular Endocrinology (CME), Ulm University, Helmholtzstrasse 8/1, 89081 Ulm, Germany; (S.V.); (S.L.); (A.-K.D.)
| |
Collapse
|
6
|
Gyanewali S, Kesharwani P, Sheikh A, Ahmad FJ, Trivedi R, Talegaonkar S. Formulation development and in vitro-in vivo assessment of protransfersomal gel of anti-resorptive drug in osteoporosis treatment. Int J Pharm 2021; 608:121060. [PMID: 34500057 DOI: 10.1016/j.ijpharm.2021.121060] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022]
Abstract
Osteoporosis is a major cause of morbidity, mortality, and economic burden worldwide. Despite being an effective in combating the bone-deteriorating disorders, bisphosphonates have several shortcomings including poor and variable bioavailability, low permeability, high toxicity, etc. In this study, we developed and optimized protransfersome formulation for the drug risedronate sodium (RIS-Na) with the goal of enhancing its bioavailability and hence patient compliance. Phase separation coacervation technique was utilized for development of optimized formulation. Optimization was achieved by using three-factor, three-level Box-Behnken design combined with Response Surface Methodology (RSM). This enabled us to decipher the effect of 3 independent variables (Phospholipid, Tween-80 and Sodium Deoxycholate) on three dependent parameters (entrapment efficiency, vesicle size and transdermal flux). Optimized formulation was further evaluated for pharmacokinetic and pharmacodynamic parameters. Smooth, spherical protransfersomes with a size of 260 ± 18 nm, having entrapment efficiency and flux of 80.4 ± 4.90% and 8.41 ± 0.148 μg/cm2/h, respectively were prepared. Ex vivo studies revealed a shorter lag time of 1.21 ± 0.18 h and higher flux associated with transdermal formulation. CLSM analysis further revealed better drug penetration (220 μm) through the skin in case of protransfersomes as compared to drug solution (72 μm). Additionally, biomechanical, biochemical, and histo-pathological studies further validated the results. Thus, it was concluded that protransfersome formulation has a great potential in providing better therapeutic efficacy of risedronate than its conventional counterpart.
Collapse
Affiliation(s)
- Suman Gyanewali
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India.
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Ritu Trivedi
- Department of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sushama Talegaonkar
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India.
| |
Collapse
|
7
|
Schündeln MM, Höppner J, Meyer FL, Schmuck W, Kauther MD, Hilken G, Levkau B, Rauner M, Grasemann C. Prednisone prevents particle induced bone loss in the calvaria mouse model. Heliyon 2021; 7:e07828. [PMID: 34471710 PMCID: PMC8387912 DOI: 10.1016/j.heliyon.2021.e07828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/03/2021] [Accepted: 08/16/2021] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Glucocorticoids are essential in the treatment of many chronic inflammatory and malignant diseases but are known to have detrimental effects on bone. This study aimed to investigate the effects of prednisone on osteoclast functioning in vivo in the calvaria particle-induced bone loss mouse model. METHODS 12-week-old male C57BL6/J mice received subcutaneously implanted prednisone (2.5 mg/d, 60 day release (n = 14)) or placebo pellets (n = 10). Osteolysis of the calvaria bone was induced two weeks later by application of ultra-high-molecular-weight polyethylene- (UHMWPE) particles to the dome (vs sham operation). The extent of osteolysis was determined histologically and by micro-computer tomography. RESULTS Prednisone significantly inhibited particle-induced osteolysis in the skull. No significant difference in osteoclast numbers was seen in mice with prednisone vs placebo treatment. Prednisone treatment alone without particle application did not reduce bone mineral density or deterioration in bone microarchitecture parameters. CONCLUSIONS The calvaria particle-induced bone loss mouse model can be adapted to investigate osteoclast activity in vivo and the effect of prednisone on osteoclasts. In this preventive experimental design, the application of short-term low-dose prednisone has osteoprotective effects without measurable systemic side effects on bone parameters.
Collapse
Affiliation(s)
- Michael M. Schündeln
- Division of Pediatric Hematology and Oncology, Department of Pediatrics III, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Jakob Höppner
- Department of Pediatrics and CeSER, Katholisches Klinikum Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Felix L. Meyer
- Department of Pediatrics II, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Wiebke Schmuck
- Department of Pediatrics II, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Max D. Kauther
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Essen, Germany
- Department for Orthopedics, Agaplesion Diakonieklinikum, Rotenburg Wümme, Germany
| | - Gero Hilken
- Central Animal Laboratory, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich-Heine-University Düsseldorf, Germany
| | - Martina Rauner
- Department of Medicine III, Dresden Technical University Medical Center, Dresden, Germany
| | - Corinna Grasemann
- Department of Pediatrics and CeSER, Katholisches Klinikum Bochum, Ruhr-University Bochum, Bochum, Germany
- Department of Pediatrics II, University Hospital Essen, University of Duisburg-Essen, Germany
| |
Collapse
|
8
|
Chen H, Song F, Long F. WNT7B overexpression rescues bone loss caused by glucocorticoids in mice. FASEB J 2021; 35:e21683. [PMID: 34118078 DOI: 10.1096/fj.202100151rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 01/28/2023]
Abstract
Glucocorticoids, widely prescribed for anti-inflammatory and immunosuppressive purposes, are the most common secondary cause for osteoporosis and related fractures. Current anti-resorptive and anabolic therapies are insufficient for treating glucocorticoid-induced osteoporosis due to contraindications or concerns of side effects. Glucocorticoids have been shown to disrupt Wnt signaling in osteoblast-lineage cells, but the efficacy for Wnt proteins to restore bone mass after glucocorticoid therapy has not been examined. Here by using two mouse genetic models wherein WNT7B expression is temporally activated by either tamoxifen or doxycycline in osteoblast-lineage cells, we show that WNT7B recovers bone mass following glucocorticoid-induced bone loss, thanks to increased osteoblast number and function. However, WNT7B overexpression in bone either before or after glucocorticoid treatments does not ameliorate the abnormal accumulation of body fat. The study demonstrates a potent bone anabolic function for WNT7B in countering glucocorticoid-induced bone loss.
Collapse
Affiliation(s)
- Hong Chen
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangfang Song
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Fanxin Long
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
9
|
Dubrovsky AM, Nyman JS, Uppuganti S, Chmiel KJ, Kimmel DB, Lane NE. Bone Strength/Bone Mass Discrepancy in Glucocorticoid-Treated Adult Mice. JBMR Plus 2020; 5:e10443. [PMID: 33778319 PMCID: PMC7990143 DOI: 10.1002/jbm4.10443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/16/2020] [Accepted: 11/22/2020] [Indexed: 11/12/2022] Open
Abstract
Glucocorticoids increase bone fragility in patients in a manner that is underestimated by bone mass measurement. This study aimed to determine if the adult mouse could model this bone strength/bone mass discrepancy. Forty‐two 13‐week‐old BALB/cJ mice were randomized into vehicle and glucocorticoid groups, implanted with vehicle or 6‐methylprednisolone pellets, and necropsied after 60 and 120 days. Bone strength and bone mass/microarchitecture were assessed at the right central femur (CF; cortical‐bone–rich) and sixth lumbar vertebral body (LVB6; trabecular‐bone–rich). Bound water (BW) of the whole right femur was analyzed by proton‐nuclear magnetic resonance (1H‐NMR) relaxometry. Data were analyzed by two‐factor ANOVA with time (day 60 and day 120) and treatment (vehicle and glucocorticoid) as main effects for all data. Significant interactions were further analyzed with a Tukey's post hoc test. Most bone strength measures in the CF were lower in the glucocorticoid group, regardless of the duration of treatment, with no time × treatment interaction. However, bone mass measures in the CF showed a significant time × treatment interaction (p = 0.0001). Bone strength measures in LVB6 showed a time × treatment interaction (p < 0.02) such that LVB6 strength was lower after 120 days of glucocorticoids compared with 120 days of vehicle treatment. Whole‐femur–BW was lower with both glucocorticoid treatment (p = 0.0001) and time (p < 0.02), with a significant time × treatment interaction (p = 0.005). Glucocorticoid treatment of male BALB/cJ mice resulted in the lowering of bone strength in both cortical and trabecular bone that either appeared earlier or was greater than the treatment‐related changes in bone mass/microarchitecture. The adult mouse may be a good model for investigating the bone strength/mass discrepancy observed in glucocorticoid‐treated patients. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alanna M Dubrovsky
- Center for Musculoskeletal Health University of California at Davis Medical Center Sacramento CA USA
| | - Jeffrey S Nyman
- Department of Orthopaedic Surgery Vanderbilt University Medical Center Nashville TN USA
| | - Sasidhar Uppuganti
- Department of Orthopaedic Surgery Vanderbilt University Medical Center Nashville TN USA
| | - Kenneth J Chmiel
- Center for Musculoskeletal Health University of California at Davis Medical Center Sacramento CA USA
| | - Donald B Kimmel
- Department of Physiological Sciences University of Florida Gainesville FL USA
| | - Nancy E Lane
- Center for Musculoskeletal Health University of California at Davis Medical Center Sacramento CA USA
| |
Collapse
|
10
|
Etani Y, Ebina K, Hirao M, Kitaguchi K, Kashii M, Ishimoto T, Nakano T, Okamura G, Miyama A, Takami K, Goshima A, Kanamoto T, Nakata K, Yoshikawa H. Combined effect of teriparatide and an anti-RANKL monoclonal antibody on bone defect regeneration in mice with glucocorticoid-induced osteoporosis. Bone 2020; 139:115525. [PMID: 32645445 DOI: 10.1016/j.bone.2020.115525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/21/2020] [Accepted: 07/02/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE The purpose of this study was to examine the effect of single or combination therapy of teriparatide (TPTD) and a monoclonal antibody against the murine receptor activator of nuclear factor κB ligand (anti-RANKL Ab) on cancellous and cortical bone regeneration in a mouse model of glucocorticoid-induced osteoporosis (GIOP). METHODS C57BL/6 J mice (24 weeks of age) were divided into five groups: (1) the SHAM group: sham operation + saline; (2) the prednisolone (PSL) group: PSL + saline; (3) the TPTD group: PSL + TPTD; (4) the Ab group: PSL + anti-RANKL Ab; and (5) the COMB group: PSL + TPTD + anti-RANKL Ab (n = 8 per group). With the exception of the SHAM group, 7.5 mg of PSL was inserted subcutaneously into mice, to generate a mouse model of GIOP. Four weeks after insertion, bone defects with a diameter of 0.9 mm were created to assess bone regeneration on both femoral metaphysis (cancellous bone) and diaphysis (cortical bone). After surgery, therapeutic intervention was continued for 4 weeks. Saline (200 μl) or TPTD (40 μg/kg) was injected subcutaneously five times per week, whereas the anti-RANKL Ab (5 mg/kg) was injected subcutaneously once on the day after surgery. Subsequently, the following analyses were performed: microstructural assessment of bone regeneration and bone mineral density (BMD) measurement via micro-computed tomography, and histological, histomorphometrical, and biomechanical analyses with nanoindentation. RESULTS The COMB group showed the highest lumbar spine BMD increase (vs. the PSL, TPTD, and Ab groups). The volume of regenerated cancellous bone at the bone defect site was higher in the COMB group compared with the PSL, TPTD, and Ab group. The volume of the regenerated cortical bone was significantly higher in the COMB group compared with the PSL group, and its hardness was significantly higher in the COMB group compared with the PSL and TPTD groups. CONCLUSION In a mouse model of glucocorticoid-induced osteoporosis, the combination therapy of TPTD plus the anti-RANKL Ab increased bone mineral density in the lumbar spine and regenerated cancellous bone volume compared with single administration of each agent, and also increased regenerated cortical bone strength compared with single administration of TPTD.
Collapse
Affiliation(s)
- Yuki Etani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | - Makoto Hirao
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kazuma Kitaguchi
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka 560-8565, Japan
| | - Masafumi Kashii
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka 560-8565, Japan
| | - Takuya Ishimoto
- Division of Materials and Manufacturing Science, Osaka University Graduate School of Engineering, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Osaka University Graduate School of Engineering, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Gensuke Okamura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Akira Miyama
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kenji Takami
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Atsushi Goshima
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takashi Kanamoto
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ken Nakata
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka 560-8565, Japan
| |
Collapse
|
11
|
Fayolle C, Labrune M, Berteau JP. Raman spectroscopy investigation shows that mineral maturity is greater in CD-1 than in C57BL/6 mice distal femurs after sexual maturity. Connect Tissue Res 2020; 61:409-419. [PMID: 30922120 DOI: 10.1080/03008207.2019.1601184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose/Aim of the study mice are the most often used pre-clinical lab models for studying the pathologies of bone mineralization. However, recent evidence suggests that two of the most often used mice strains (C57BL/6J and CD-1) might show differences in the bone mineralization process. This study sought to investigate the main compositional properties of bone tissue between nonpathological C57BL/6J and CD-1 murine knee joints. Materials and Methods : to this end, medial and lateral condylar subchondral bones and the adjacent diaphyseal cortical bone of 13 murine femurs (n = 7 C57BL/6J and n = 6 CD-1 at eight weeks old, just after sexual maturation) were analyzed with ex vivo Raman spectroscopy. Results : regardless of the bone tissue analyzed, our results showed that CD-1 laboratory mice present a more mature mineral phase than C57BL/6J laboratory mice, but present no difference in maturity of the collagen phase. For both strains, the subchondral bone of the medial condylar and cortical bone from the diaphysis have similar compositional properties, and CD-1 presents less variation than C57BL/6J. Furthermore, we depict a novel parametric relationship between the crystallinity and carbonate-to-amide-I ratio that might help in deciphering the mineral maturation process that occurs during bone's mineralization. Conclusions : Our results suggest that the timing of bone maturation might be different between non-pathological C57BL/6J and CD-1 murine knee femurs.
Collapse
Affiliation(s)
- Clémence Fayolle
- Department of Physical Therapy, City University of New York, College of Staten Island , New York, NY, USA.,Department of Biomedical Engineering, Compiegne, Sorbonne University, Universite Technologique de Compiegne , France
| | - Mélody Labrune
- Department of Physical Therapy, City University of New York, College of Staten Island , New York, NY, USA.,Department of Biomedical Engineering, Compiegne, Sorbonne University, Universite Technologique de Compiegne , France
| | - Jean-Philippe Berteau
- Department of Physical Therapy, City University of New York, College of Staten Island , New York, NY, USA.,New York Center for Biomedical Engineering, City University of New York, City College , New York, NY, USA.,Nanoscience Initiatives, Advanced Science Research Center, City University of New York, City College , New York, NY, USA
| |
Collapse
|
12
|
Yun B, Maburutse BE, Kang M, Park MR, Park DJ, Kim Y, Oh S. Short communication: Dietary bovine milk-derived exosomes improve bone health in an osteoporosis-induced mouse model. J Dairy Sci 2020; 103:7752-7760. [PMID: 32622594 DOI: 10.3168/jds.2019-17501] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Osteoporosis is a systemic skeletal disease characterized by low bone mass and micro-architectural deterioration of bone tissue, with a consequent increase in bone fragility and fracture susceptibility. In an aged society with increased life expectancy, the incidence rate of osteoporosis is also rapidly increasing. Inadequate nutrition may negatively influence bone metabolism. Recently, many studies have investigated the functionality of milk-derived exosomes, which play important roles in cell-to-cell communication. However, there are few reports of how milk-derived exosomes influence osteoblast proliferation and differentiation. Here, we determined whether bovine colostrum-derived exosomes promote anti-osteoporosis in vitro and in vivo. Tartrate-resistant acid phosphatase-stained cells were significantly inhibited in Raw264.7 cells treated with exosomes, indicating reduced osteoclast differentiation. We induced osteoporosis in mice using glucocorticoid pellets after orally administering exosomes for 2 mo. Interestingly, the bone mineral density of exosome-fed mouse groups was significantly improved compared with the glucocorticoid-induced osteoporosis group without exosome treatment. In addition, Lactobacillus were decreased in the gut microbiota community of osteoporosis-induced mice, but the gut microbiota community composition was effectively restored by exosome intake. Taken together, we propose that exosomes isolated from bovine colostrum could be a potential candidate for osteoporosis prevention, bone remodeling improvement, and inhibition of bone resorption. To our knowledge, this is the first time that a protective effect of milk exosomes against osteoporosis has been demonstrated in vivo. Our results strongly suggest that bovine colostrum exosomes might be used as a prophylaxis to prevent the onset of osteoporosis. Indeed, our results offer promising alternative strategies in the nutritional management of age-related bone complications.
Collapse
Affiliation(s)
- B Yun
- Department of Animal Science and Institute of Milk Genomics, Chonbuk National University, Jeonju 54896, Korea
| | - B E Maburutse
- Department of Animal Sciences and Health, Marondera University of Agricultural Sciences and Health, PO Box 35, Marondera, Zimbabwe
| | - M Kang
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju 55069, Korea
| | - M R Park
- Department of Pharmacology and System Physiology, University of Cincinnati, OH 45267
| | - D J Park
- Korea Food Research Institute, Jeollabuk-do 55365, Korea
| | - Y Kim
- Department of Agricultural Biotechnology and Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea.
| | - S Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju 55069, Korea.
| |
Collapse
|
13
|
Sato D, Takahata M, Ota M, Fukuda C, Hasegawa T, Yamamoto T, Amizuka N, Tsuda E, Okada A, Hiruma Y, Fujita R, Iwasaki N. Siglec-15-targeting therapy protects against glucocorticoid-induced osteoporosis of growing skeleton in juvenile rats. Bone 2020; 135:115331. [PMID: 32217159 DOI: 10.1016/j.bone.2020.115331] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
Abstract
Effective treatment of juvenile osteoporosis, which is frequently caused by glucocorticoid (GC) therapy, has not been established due to limited data regarding the efficacy and adverse effects of antiresorptive therapies on the growing skeleton. We previously demonstrated that sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) targeting therapy, which interferes with osteoclast terminal differentiation in the secondary, but not primary, spongiosa, increased bone mass without adverse effects on skeletal growth, whereas bisphosphonate, a first-line treatment for osteoporosis, increased bone mass but impaired long bone growth in healthy growing rats. In the present study, we investigated the efficacy of anti-Siglec-15 neutralizing antibody (Ab) therapy against GC-induced osteoporosis in a growing rat model. GC decreased bone mass and deteriorated mechanical properties of bone, due to a disproportionate increase in bone resorption. Both anti-Siglec-15 Ab and alendronate (ALN) showed protective effects against GC-induced bone loss by suppressing bone resorption, which was more pronounced with anti-Siglec-15 Ab treatment, possibly due to a reduced negative impact on bone formation. ALN induced histological abnormalities in the growth plate and morphological abnormalities in the long bone metaphysis but did not cause significant growth retardation. Conversely, anti-Siglec-15 Ab did not show any negative impact on the growth plate and preserved normal osteoclast and chondroclast function at the primary spongiosa. Taken together, these results suggest that anti-Siglec-15 targeting therapy could be a safe and efficacious prophylactic therapy for GC-induced osteoporosis in juvenile patients.
Collapse
Affiliation(s)
- Dai Sato
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masahiko Takahata
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| | - Masahiro Ota
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Chie Fukuda
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Tomoka Hasegawa
- Hokkaido University, Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Sapporo, Japan
| | - Tomomaya Yamamoto
- Hokkaido University, Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Sapporo, Japan
| | - Norio Amizuka
- Hokkaido University, Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Sapporo, Japan
| | - Eisuke Tsuda
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Akiko Okada
- Specialty Medicine Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yoshiharu Hiruma
- Pharmacovigilance Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Ryo Fujita
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
14
|
Jiang Y, Lu Y, Jiang X, Hu J, Li R, Liu Y, Zhu G, Rong X. Glucocorticoids induce osteoporosis mediated by glucocorticoid receptor-dependent and -independent pathways. Biomed Pharmacother 2020; 125:109979. [PMID: 32044718 DOI: 10.1016/j.biopha.2020.109979] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/18/2020] [Accepted: 01/27/2020] [Indexed: 12/30/2022] Open
Abstract
Clinically, glucocorticoids (GCs) are widely used to treat inflammation-related diseases; however, their long-term use causes side effects, such as osteoporosis and predisposition to bone fractures, known as glucocorticoid-induced osteoporosis (GIOP). Nr3c1 is the major glucocorticoid receptor, and its downstream signaling pathway is involved in regulating various intracellular physiological processes, including those related to bone cells; however, its mechanism in glucocorticoid-induced osteoporosis (GIOP) remains unclear. In this study, a zebrafish nr3c1-mutant was successfully generated using CRISPR/Cas9 technology to investigate the role of nr3c1 in GIOP. Mutations in nr3c1 altered cartilage development and significantly decreased bone mineralization area. Additionally, qRT-PCR results showed that the expression of extracellular matrix-, osteoblast-, and osteoclast-related genes was altered in the nr3c1-mutant. The GC-Nr3c1 pathway regulates the expression of extracellular matrix-, osteoblast-, and osteoclast-related genes via Nr3c1-dependent and Nr3c1-independent pathways. A dual-luciferase reporter assay further revealed that GCs and Nr3c1 transcriptionally regulate matrix metalloproteinase 9 (mmp9), alkaline phosphatase (alp), and acid phosphatase 5a (acp5a). This study reveals that GCs/Nr3c1 affect the expression of genes involved in bone metabolism and provides a basis to determine the role of GIOP and Nr3c1 in bone metabolism and development. We also identified a new effector target for the clinical treatment of GIOP.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Orthopedics, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Yajun Lu
- Department of Orthopedics, Yixin Shanjuan Orthopaedic Hospital, YiXing, Jiangsu, 214000, China
| | - Xu Jiang
- Department of Orthopedics, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Jiawei Hu
- Department of Orthopedics, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Rong Li
- Department of Pharmacy, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Yun Liu
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350 China
| | - Guoxing Zhu
- Department of Orthopedics, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, China.
| | - Xiaoxu Rong
- Department of Orthopedics, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, China.
| |
Collapse
|
15
|
Okada K, Okamoto T, Okumoto K, Takafuji Y, Ishida M, Kawao N, Matsuo O, Kaji H. PAI-1 is involved in delayed bone repair induced by glucocorticoids in mice. Bone 2020; 134:115310. [PMID: 32142912 DOI: 10.1016/j.bone.2020.115310] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/21/2020] [Accepted: 03/02/2020] [Indexed: 02/05/2023]
Abstract
Glucocorticoid (GC) treatments induce osteoporosis and chronic GC treatments have been suggested to induce delayed bone repair; however, the mechanisms by which GC induces delayed bone repair remain unclear. We herein investigated the roles of plasminogen activator inhibitor-1 (PAI-1) in GC-induced effects on bone repair after femoral bone injury using female mice with a PAI-1 deficiency and their wild-type counterparts. Dexamethasone (Dex) increased plasma PAI-1 levels as well as PAI-1 mRNA levels in the adipose tissues and muscles of wild-type mice. PAI-1 deficiency significantly blunted Dex-induced delayed bone repair in mice. Moreover, PAI-1 deficiency significantly blunted Runx2 mRNA levels suppressed by Dex as well as Dex-induced osteoblast apoptosis at the damaged site 7 days after bone injury in mice. On the other hand, PAI-1 deficiency did not affect adipogenic gene expression enhanced by Dex at the damaged site 7 days after bone injury in mice. In conclusion, we herein showed for the first time that PAI-1 is involved in delayed bone repair after bone injury induced by GC in mice. PAI-1 may influence early stage osteoblast differentiation and apoptosis during the osteoblastic restoration phase of the bone repair process.
Collapse
Affiliation(s)
- Kiyotaka Okada
- Department of Arts and Science, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan; Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Takahiro Okamoto
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Katsumi Okumoto
- Life Science Research Institute, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yoshimasa Takafuji
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masayoshi Ishida
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Osamu Matsuo
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan.
| |
Collapse
|
16
|
Schepper JD, Collins F, Rios-Arce ND, Kang HJ, Schaefer L, Gardinier JD, Raghuvanshi R, Quinn RA, Britton R, Parameswaran N, McCabe LR. Involvement of the Gut Microbiota and Barrier Function in Glucocorticoid-Induced Osteoporosis. J Bone Miner Res 2020; 35:801-820. [PMID: 31886921 DOI: 10.1002/jbmr.3947] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/05/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022]
Abstract
Glucocorticoids (GCs) are potent immune-modulating drugs with significant side effects, including glucocorticoid-induced osteoporosis (GIO). GCs directly induce osteoblast and osteocyte apoptosis but also alter intestinal microbiota composition. Although the gut microbiota is known to contribute to the regulation of bone density, its role in GIO has never been examined. To test this, male C57/Bl6J mice were treated for 8 weeks with GC (prednisolone, GC-Tx) in the presence or absence of broad-spectrum antibiotic treatment (ABX) to deplete the microbiota. Long-term ABX prevented GC-Tx-induced trabecular bone loss, showing the requirement of gut microbiota for GIO. Treatment of GC-Tx mice with a probiotic (Lactobacillus reuteri [LR]) prevented trabecular bone loss. Microbiota analyses indicated that GC-Tx changed the abundance of Verrucomicobiales and Bacteriodales phyla and random forest analyses indicated significant differences in abundance of Porphyromonadaceae and Clostridiales operational taxonomic units (OTUs) between groups. Furthermore, transplantation of GC-Tx mouse fecal material into recipient naïve, untreated WT mice caused bone loss, supporting a functional role for microbiota in GIO. We also report that GC caused intestinal barrier breaks, as evidenced by increased serum endotoxin level (2.4-fold), that were prevented by LR and ABX treatments. Enhancement of barrier function with a mucus supplement prevented both GC-Tx-induced barrier leakage and trabecular GIO. In bone, treatment with ABX, LR or a mucus supplement reduced GC-Tx-induced osteoblast and osteocyte apoptosis. GC-Tx suppression of Wnt10b in bone was restored by the LR and high-molecular-weight polymer (MDY) treatments as well as microbiota depletion. Finally, we identified that bone-specific Wnt10b overexpression prevented GIO. Taken together, our data highlight the previously unappreciated involvement of the gut microbiota and intestinal barrier function in trabecular GIO pathogenesis (including Wnt10b suppression and osteoblast and osteocyte apoptosis) and identify the gut as a novel therapeutic target for preventing GIO. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Fraser Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, MI, USA.,Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MI, USA
| | - Ho Jun Kang
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Laura Schaefer
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Ruma Raghuvanshi
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Robert Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
17
|
Fenton CG, Doig CL, Fareed S, Naylor A, Morrell AP, Addison O, Wehmeyer C, Buckley CD, Cooper MS, Lavery GG, Raza K, Hardy RS. 11β-HSD1 plays a critical role in trabecular bone loss associated with systemic glucocorticoid therapy. Arthritis Res Ther 2019; 21:188. [PMID: 31420008 PMCID: PMC6698000 DOI: 10.1186/s13075-019-1972-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 08/07/2019] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Despite their efficacy in the treatment of chronic inflammation, the prolonged application of therapeutic glucocorticoids (GCs) is limited by significant systemic side effects including glucocorticoid-induced osteoporosis (GIOP). 11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) is a bi-directional enzyme that primarily activates GCs in vivo, regulating tissue-specific exposure to active GC. We aimed to determine the contribution of 11β-HSD1 to GIOP. METHODS Wild type (WT) and 11β-HSD1 knockout (KO) mice were treated with corticosterone (100 μg/ml, 0.66% ethanol) or vehicle (0.66% ethanol) in drinking water over 4 weeks (six animals per group). Bone parameters were assessed by micro-CT, sub-micron absorption tomography and serum markers of bone metabolism. Osteoblast and osteoclast gene expression was assessed by quantitative RT-PCR. RESULTS Wild type mice receiving corticosterone developed marked trabecular bone loss with reduced bone volume to tissue volume (BV/TV), trabecular thickness (Tb.Th) and trabecular number (Tb.N). Histomorphometric analysis revealed a dramatic reduction in osteoblast numbers. This was matched by a significant reduction in the serum marker of osteoblast bone formation P1NP and gene expression of the osteoblast markers Alp and Bglap. In contrast, 11β-HSD1 KO mice receiving corticosterone demonstrated almost complete protection from trabecular bone loss, with partial protection from the decrease in osteoblast numbers and markers of bone formation relative to WT counterparts receiving corticosterone. CONCLUSIONS This study demonstrates that 11β-HSD1 plays a critical role in GIOP, mediating GC suppression of anabolic bone formation and reduced bone volume secondary to a decrease in osteoblast numbers. This raises the intriguing possibility that therapeutic inhibitors of 11β-HSD1 may be effective in preventing GIOP in patients receiving therapeutic steroids.
Collapse
Affiliation(s)
- C. G. Fenton
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - C. L. Doig
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - S. Fareed
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - A. Naylor
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - A. P. Morrell
- Aston Institute of Materials Research, Aston University, Birmingham, UK
| | - O. Addison
- Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - C. Wehmeyer
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - C. D. Buckley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - M. S. Cooper
- ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - G. G. Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - K. Raza
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | - R. S. Hardy
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
18
|
Ohlsson C, Nilsson KH, Henning P, Wu J, Gustafsson KL, Poutanen M, Lerner UH, Movérare-Skrtic S. WNT16 overexpression partly protects against glucocorticoid-induced bone loss. Am J Physiol Endocrinol Metab 2018; 314:E597-E604. [PMID: 29406783 DOI: 10.1152/ajpendo.00292.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapeutic use of glucocorticoids (GCs) is a major cause of secondary osteoporosis, but the molecular mechanisms responsible for the deleterious effects of GCs in bone are only partially understood. WNT16 is a crucial physiological regulator of bone mass and fracture susceptibility, and we hypothesize that disturbed WNT16 activity might be involved in the deleterious effects of GC in bone. Twelve-week-old female Obl-Wnt16 mice (WNT16 expression driven by the rat procollagen type I α1 promoter) and wild-type (WT) littermates were treated with prednisolone (7.6 mg·kg-1·day-1) or vehicle for 4 wk. We first observed that GC treatment decreased the Wnt16 mRNA levels in bone of female mice (-56.4 ± 6.1% compared with vehicle, P < 0.001). We next evaluated if WNT16 overexpression protects against GC-induced bone loss. Dual-energy X-ray absorptiometry analyses revealed that GC treatment decreased total body bone mineral density in WT mice (-3.9 ± 1.2%, P = 0.028) but not in Obl-Wnt16 mice (+1.3 ± 1.4%, nonsignificant). Microcomputed tomography analyses showed that GC treatment decreased trabecular bone volume fraction (BV/TV) of the femur in WT mice ( P = 0.019) but not in Obl-Wnt16 mice. Serum levels of the bone formation marker procollagen type I N-terminal propeptide were substantially reduced by GC treatment in WT mice (-50.3 ± 7.0%, P = 0.008) but not in Obl-Wnt16 mice (-3.8 ± 21.2%, nonsignificant). However, the cortical bone thickness in femur was reduced by GC treatment in both WT mice and Obl-Wnt16 mice. In conclusion, GC treatment decreases Wnt16 mRNA levels in bone and WNT16 overexpression partly protects against GC-induced bone loss.
Collapse
Affiliation(s)
- Claes Ohlsson
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Karin H Nilsson
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Petra Henning
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Jianyao Wu
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Karin L Gustafsson
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Matti Poutanen
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
- Institute of Biomedicine and Turku Center for Disease Modeling, University of Turku , Turku , Finland
| | - Ulf H Lerner
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Sofia Movérare-Skrtic
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
19
|
Huang H, Lin H, Lan F, Wu Y, Yang Z, Zhang J. Application of bone transgenic zebrafish in anti-osteoporosis chemical screening. Animal Model Exp Med 2018; 1:53-61. [PMID: 30891547 PMCID: PMC6354313 DOI: 10.1002/ame2.12000] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/16/2018] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The zebrafish (Danio rerio) has recently been shown to be an ideal model to study bone disease including osteoporosis. The zebrafish osteoporosis model could be induced by glucocorticoid treatment with chemical staining for reflecting the level of bone mineralization. However, this methodology was unstable. Here, we developed a novel methodology to directly evaluate the bone mass and density. METHODS We generated and used the bone of transgenic zebrafish Tg (ola.sp7:nlsGFP) to evaluate the bone mass and density by measuring the areal extent and the integrated optical density (IOD) of enhanced green fluorescent protein (eGFP). This methodology was further compared with the traditional chemically stained method showing the bone mineralization. Furthermore, genes related to zebrafish osteoporosis were analyzed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). RESULTS Our results of new methods were consistent with those from chemically stained fish, following glucocorticoid-induction or epimedium flavonoid (FE)-rescue treatments. qRT-PCR analyses on mRNA levels revealed that glucocorticoid induces osteoporosis by downregulating the expression of osteoblast-related factors osterix, osteocalcin, and osteopontin, and upregulating the expression of osteoclast-related factor tartrate-resistant acid phosphatase. In FE-rescued fish, the expression of osteogenic factors osterix, osteocalcin, and osteopontin were increased. CONCLUSION Compared to the traditional chemical staining methods, the new osteoporosis model using Tg(ola.sp7:nlsGFP) is more convenient and efficient for studying osteoporosis in vivo, and especially for high-throughput anti-osteoporosis drug screening.
Collapse
Affiliation(s)
- Hong‐xin Huang
- Affiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvinceChina
| | - Hao Lin
- Affiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvinceChina
| | - Fen Lan
- The Central People's Hospital of HuizhouGuangdongGuangdong ProvinceChina
| | - Yong‐fu Wu
- Affiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvinceChina
| | - Zhen‐guo Yang
- Affiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvinceChina
| | - Jing‐jing Zhang
- Affiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong ProvinceChina
| |
Collapse
|
20
|
Li C, Chai Y, Wang L, Gao B, Chen H, Gao P, Zhou FQ, Luo X, Crane JL, Yu B, Cao X, Wan M. Programmed cell senescence in skeleton during late puberty. Nat Commun 2017; 8:1312. [PMID: 29101351 PMCID: PMC5670205 DOI: 10.1038/s41467-017-01509-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/22/2017] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem/progenitor cells (MSPCs) undergo rapid self-renewal and differentiation, contributing to fast skeletal growth during childhood and puberty. It remains unclear whether these cells change their properties during late puberty to young adulthood, when bone growth and accrual decelerate. Here we show that MSPCs in primary spongiosa of long bone in mice at late puberty undergo normal programmed senescence, characterized by loss of nestin expression. MSPC senescence is epigenetically controlled by the polycomb histone methyltransferase enhancer of zeste homolog 2 (Ezh2) and its trimethylation of histone H3 on Lysine 27 (H3K27me3) mark. Ezh2 maintains the repression of key cell senescence inducer genes through H3K27me3, and deletion of Ezh2 in early pubertal mice results in premature cellular senescence, depleted MSPCs pool, and impaired osteogenesis as well as osteoporosis in later life. Our data reveals a programmed cell fate change in postnatal skeleton and unravels a regulatory mechanism underlying this phenomenon. Mesenchymal stem cells are essential for bone development, but it is unclear if their activity is maintained after late puberty, when bone growth decelerates. The authors show that during late puberty in mice, these cells undergo senescence under the epigenetic control of Ezh2.
Collapse
Affiliation(s)
- Changjun Li
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu Chai
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Bo Gao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hao Chen
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Peisong Gao
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Janet L Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Bin Yu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
21
|
Williams-Dautovich J, Yogendirarajah K, Dela Cruz A, Patel R, Tsai R, Morgan SA, Mitchell J, Grynpas MD, Cummins CL. The CRH-Transgenic Cushingoid Mouse Is a Model of Glucocorticoid-Induced Osteoporosis. JBMR Plus 2017; 1:46-57. [PMID: 30283880 PMCID: PMC6124163 DOI: 10.1002/jbm4.10009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/23/2017] [Accepted: 05/28/2017] [Indexed: 12/21/2022] Open
Abstract
Glucocorticoids (GCs) have unparalleled anti‐inflammatory and immunosuppressive properties, which accounts for their widespread prescription and use. Unfortunately, a limitation to GC therapy is a wide range of negative side effects including Cushing's syndrome, a disease characterized by metabolic abnormalities including muscle wasting and osteoporosis. GC‐induced osteoporosis occurs in 30% to 50% of patients on GC therapy and thus, represents an important area of study. Herein, we characterize the molecular and physiologic effects of GC‐induced osteoporosis using the Cushing's mouse model, the corticotropin releasing hormone (CRH) transgenic mouse (CRH‐Tg). The humeri, femurs, and tibias from wild‐type (WT) and CRH‐Tg male mice, aged 13 to 14 weeks old were subjected to multiple bone tests including, micro–computed tomography (μCT), static and dynamic histomorphometry, strength testing, and gene expression analyses. The CRH‐Tg mice had a 38% decrease in cortical bone area, a 35% decrease in cortical thickness, a 16% decrease in trabecular thickness, a sixfold increase in bone adiposity, a 27% reduction in osteoid width, a 75% increase in bone‐resorbing osteoclast number/bone surface, a 34% decrease in bone formation rate, and a 40% decrease in bone strength compared to WT mice. At the gene expression level, CRH‐Tg bone showed significantly increased osteoclast markers and decreased osteoblast markers, whereas CRH‐Tg muscle had increased muscle atrophy gene markers compared to WT mice. Overall, the CRH‐Tg mouse model aged to 14 weeks recapitulated many features of osteoporosis in Cushing's syndrome and thus, represents a useful model to study GC‐induced osteoporosis and interventions that target the effects of GCs on the skeleton. © 2017 The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | | | - Ariana Dela Cruz
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
| | - Rucha Patel
- Department of Pharmaceutical Sciences University of Toronto Toronto Ontario Canada
| | - Ricky Tsai
- Department of Pharmaceutical Sciences University of Toronto Toronto Ontario Canada
| | - Stuart A Morgan
- Department of Pharmaceutical Sciences University of Toronto Toronto Ontario Canada
| | - Jane Mitchell
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
| | - Marc D Grynpas
- Lunenfeld-Tanenbaum Research Institute Mount Sinai Hospital Toronto Ontario Canada.,Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences University of Toronto Toronto Ontario Canada
| |
Collapse
|
22
|
Strain dependent differences in glucocorticoid-induced bone loss between C57BL/6J and CD-1 mice. Sci Rep 2016; 6:36513. [PMID: 27812009 PMCID: PMC5109915 DOI: 10.1038/srep36513] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/30/2016] [Indexed: 01/08/2023] Open
Abstract
We have investigated the effect of long-term glucocorticoid (GC) administration on bone turnover in two frequently used mouse strains; C57BL/6J and CD1, in order to assess the influence of their genetic background on GC-induced osteoporosis (GIO). GIO was induced in 12 weeks old female C57BL/6J and CD1 mice by subcutaneous insertion of long-term release prednisolone or placebo pellets. Biomechanical properties as assessed by three point bent testing revealed that femoral elasticity and strength significantly decreased in CD1 mice receiving GC, whereas C57BL/6J mice showed no differences between placebo and prednisolone treatment. Bone turnover assessed by microcomputer tomography revealed that contrary to C57BL/6J mice, prednisolone treated CD1 mice developed osteoporosis. In vitro experiments have underlined that, at a cellular level, C57BL/6J mice osteoclasts and osteoblasts were less responsive to GC treatment and tolerated higher doses than CD1 cells. Whilst administration of long-term release prednisolone pellets provided a robust GIO animal model in 12 weeks old CD1 mice, age matched C57BL/6J mice were not susceptible to the bone changes associated with GIO. This study indicates that for the induction of experimental GIO, the mouse strain choice together with other factors such as age should be carefully evaluated.
Collapse
|
23
|
Baschant U, Henneicke H, Hofbauer LC, Rauner M. Sclerostin Blockade-A Dual Mode of Action After All? J Bone Miner Res 2016; 31:1787-1790. [PMID: 27597566 DOI: 10.1002/jbmr.2988] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/25/2016] [Accepted: 09/01/2016] [Indexed: 12/24/2022]
Affiliation(s)
- Ulrike Baschant
- Department of Medicine 3, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Holger Henneicke
- Department of Medicine 3, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.,Deutsche Forschungsgemeinschaft (DFG)-Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine 3, Technische Universität Dresden, Dresden, Germany. .,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany. .,Deutsche Forschungsgemeinschaft (DFG)-Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany.
| | - Martina Rauner
- Department of Medicine 3, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
24
|
Wu Q, Xiong X, Zhang X, Lu J, Zhang X, Chen W, Wu T, Cui L, Liu Y, Xu B. Secondary osteoporosis in collagen-induced arthritis rats. J Bone Miner Metab 2016. [PMID: 26210858 DOI: 10.1007/s00774-015-0700-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Numerous studies have demonstrated that rheumatoid arthritis (RA) is often associated with bone loss; however, few experiments have focused on cancellous and cortical bone changes in rats during the process of arthritis. We have investigated bone changes in rats with collagen-induced arthritis (CIA) and have explored the characteristics of how RA induces osteoporosis by means of bone histomorphometry, bone biomechanics studies, bone mineral density studies, micro computer tomography, enzyme-linked immunosorbant assay, immunohistochemistry, and Western blot analysis. Bone mineral density of the femur and lumbar vertebrae and biomechanical properties of the femur were decreased in CIA rats. Trabecular bone volume of the tibia and lumbar vertebrae was decreased whereas bone resorption was increased in CIA rats. Bone formation of the tibial shaft in periosteal surfaces was decreased in CIA rats. Furthermore, the trabecular bone loss in CIA rats was severer at 16 weeks than at 8 weeks, as was cortical bone loss. The serum level of tumor necrosis factor α in CIA rats was increased, and the expression of dickkopf 1 and that of receptor activator of nuclear factor κB (RANKL) ligand (RANKL) in the ankle joints were also increased, but the expression of osteoprotegerin (OPG) was decreased. We conclude that CIA rats developed systemic osteoporosis, and that osteoporosis became more serious with CIA development. The mechanism may be related to the increase of bone resorption in cancellous bone cause by upregulation of the expression of DKK-1 and regulation of the RANKL/RANK/OPG signaling pathway, and the decrease of bone formation in cortical bone caused by an increase in the expression of DKK-1.
Collapse
Affiliation(s)
- Qingyun Wu
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Xueting Xiong
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Xinle Zhang
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Jiaqi Lu
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Xuemei Zhang
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Wenshuang Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Tie Wu
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Liao Cui
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Yuyu Liu
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China
| | - Bilian Xu
- Department of Pharmacology, Guangdong Medical University, No. 2, Wenming Donglu, Xiashan District, Zhanjiang, 524023, Guangdong, People's Republic of China.
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, 524023, Guangdong, People's Republic of China.
| |
Collapse
|