51
|
Baraghithy S, Smoum R, Attar-Namdar M, Mechoulam R, Bab I, Tam J. HU-671, a Novel Oleoyl Serine Derivative, Exhibits Enhanced Efficacy in Reversing Ovariectomy-Induced Osteoporosis and Bone Marrow Adiposity. Molecules 2019; 24:molecules24203719. [PMID: 31623098 PMCID: PMC6832161 DOI: 10.3390/molecules24203719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/05/2019] [Accepted: 10/15/2019] [Indexed: 01/06/2023] Open
Abstract
Oleoyl serine (OS), an endogenous fatty acyl amide (FAA) found in bone, has been shown to have an anti-osteoporotic effect. OS, being an amide, can be hydrolyzed in the body by amidases. Hindering its amide bond by introducing adjacent substituents has been demonstrated as a successful method for prolonging its skeletal activity. Here, we tested the therapeutic efficacy of two methylated OS derivatives, oleoyl α-methyl serine (HU-671) and 2-methyl-oleoyl serine (HU-681), in an ovariectomized mouse model for osteoporosis by utilizing combined micro-computed tomography, histomorphometry, and cell culture analyses. Our findings indicate that daily treatment for 6 weeks with OS or HU-671 completely rescues bone loss, whereas HU-681 has only a partial effect. The increased bone density was primarily due to enhanced trabecular thickness and number. Moreover, the most effective dose of HU-671 was 0.5 mg/kg/day, an order of magnitude lower than with OS. The reversal of bone loss resulted from increased bone formation and decreased bone resorption, as well as reversal of bone marrow adiposity. These results were further confirmed by determining the serum levels of osteocalcin and type 1 collagen C-terminal crosslinks, as well as demonstrating the enhanced antiadipogenic effect of HU-671. Taken together, these data suggest that methylation interferes with OS’s metabolism, thus enhancing its effects by extending its availability to its target cells.
Collapse
Affiliation(s)
- Saja Baraghithy
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
- Bone Laboratory, Institute for Dental Research, Faculty of Dentistry, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (R.S.)
| | - Reem Smoum
- Bone Laboratory, Institute for Dental Research, Faculty of Dentistry, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (R.S.)
- Medicinal Chemistry Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Malka Attar-Namdar
- Bone Laboratory, Institute for Dental Research, Faculty of Dentistry, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (R.S.)
| | - Raphael Mechoulam
- Medicinal Chemistry Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Itai Bab
- Bone Laboratory, Institute for Dental Research, Faculty of Dentistry, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (R.S.)
| | - Joseph Tam
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
- Correspondence: ; Tel.: +972-2-6757645; Fax: +972-2-6757015
| |
Collapse
|
52
|
Shen GY, Ren H, Shang Q, Zhao WH, Zhang ZD, Yu X, Huang JJ, Tang JJ, Yang ZD, Liang D, Jiang XB. Let-7f-5p regulates TGFBR1 in glucocorticoid-inhibited osteoblast differentiation and ameliorates glucocorticoid-induced bone loss. Int J Biol Sci 2019; 15:2182-2197. [PMID: 31592234 PMCID: PMC6775285 DOI: 10.7150/ijbs.33490] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/11/2019] [Indexed: 12/21/2022] Open
Abstract
Previous studies indicated that let-7 enhances osteogenesis and bone formation of human adipose-derived mesenchymal stem cells (MSCs). We also have confirmed that let-7f-5p expression was upregulated during osteoblast differentiation in rat bone marrow-derived MSCs (BMSCs) and was downregulated in the vertebrae of patients with glucocorticoid (GC)-induced osteoporosis (GIOP). The study was performed to determine the role of let-7f-5p in GC-inhibited osteogenic differentiation of murine BMSCs in vitro and in GIOP in vivo. Here, we report that dexamethasone (Dex) inhibited osteogenic differentiation of BMSCs and let-7f-5p expression, while increasing the expression of transforming growth factor beta receptor 1 (TGFBR1), a direct target of let-7f-5p during osteoblast differentiation under Dex conditions. In addition, let-7f-5p promoted osteogenic differentiation of BMSCs, as indicated by the promotion of alkaline phosphatase (ALP) staining and activity, Von Kossa staining, and osteogenic marker expression (Runx2,Osx, Alp, and Ocn), but decreased TGFBR1 expression in the presence of Dex. However, overexpression of TGFBR1 reversed the upregulation of let-7f-5p during Dex-treated osteoblast differentiation. Knockdown of TGFBR1 reversed the effect of let-7f-5p downregulation during Dex-treated osteogenic differentiation of BMSCs. We also found that glucocorticoid receptor (GR) mediated transcriptional silencing of let-7f-5p and its knockdown enhanced Dex-inhibited osteogenic differentiation. Further, when injected in vivo, agomiR-let-7f-5p significantly reversed bone loss induced by Dex, as well as increased osteogenic marker expression (Runx2, Osx, Alp, and Ocn) and decreased TGFBR1 expression in bone extracts. These findings indicated that the regulatory axis of GR/let-7f-5p/TGFBR1 may be important for Dex-inhibited osteoblast differentiation and that let-7f-5p may be a useful therapeutic target for GIOP.
Collapse
Affiliation(s)
- Geng-Yang Shen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hui Ren
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wen-Hua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhi-Da Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jin-Jing Huang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jing-Jing Tang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhi-Dong Yang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiao-Bing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
53
|
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: 4.8] [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
|
54
|
Porwal K, Pal S, Tewari D, Pal China S, Singh P, Chandra Tewari M, Prajapati G, Singh P, Cheruvu S, Khan YA, Sanyal S, Gayen JR, Ampapathi R, Mridha AR, Chattopadhyay N. Increased Bone Marrow-Specific Adipogenesis by Clofazimine Causes Impaired Fracture Healing, Osteopenia, and Osteonecrosis Without Extraskeletal Effects in Rats. Toxicol Sci 2019; 172:167-180. [PMID: 31393584 DOI: 10.1093/toxsci/kfz172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022] Open
Abstract
AbstractMycobacterium leprae infection causes bone lesions and osteoporosis, however, the effect of antileprosy drugs on the bone is unknown. We, therefore, set out to address it by investigating osteogenic differentiation from bone marrow (BM)-derived mesenchymal stem cells (MSCs). Out of 7 antileprosy drugs, only clofazimine (CFZ) reduced MSCs viability (IC50 ∼ 1 μM) and their osteogenic differentiation but increased adipogenic differentiation on a par with rosiglitazone, and this effect was blocked by a peroxisome proliferator-activated receptor gamma antagonist, GW9662. CFZ also decreased osteoblast viability and resulted in impaired bone regeneration in a rat femur osteotomy model at one-third human drug dose owing to increased callus adipogenesis as GW9662 prevented this effect. CFZ treatment decreased BM MSC population and homing of MSC to osteotomy site despite drug levels in BM being much less than its in vitro IC50 value. In adult rats, CFZ caused osteopenia in long bones marked by suppressed osteoblast function due to enhanced adipogenesis and increased osteoclast functions. A robust increase in marrow adipose tissue (MAT) by CFZ did not alter the hematologic parameters but likely reduced BM vascular bed leading to osteonecrosis (ON) characterized by empty osteocyte lacunae. However, CFZ had no effect on visceral fat content and was not associated with any metabolic and hematologic changes. Levels of unsaturated fatty acids in MAT were higher than saturated fatty acids and CFZ further increased the former. From these data, we conclude that CFZ has adverse skeletal effects and could be used for creating a rodent ON model devoid of extraskeletal effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sabyasachi Sanyal
- Division of Biochemistry, CSIR-Central Drug Research Institute, Lucknow 226 031, India
| | | | | | - Asit R Mridha
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110023, India
| | | |
Collapse
|
55
|
Delivanis DA, Athimulam S, Bancos I. Modern Management of Mild Autonomous Cortisol Secretion. Clin Pharmacol Ther 2019; 106:1209-1221. [PMID: 31206616 DOI: 10.1002/cpt.1551] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022]
Abstract
Incidentally discovered adrenal tumors are reported in ~ 5% of adults undergoing cross-sectional imaging. Mild autonomous cortisol secretion (MACS) from the adrenal mass is demonstrated in 5-48% of patients with adrenal tumors. The diagnosis of MACS represents a challenge due to limitations of the currently used diagnostic tests, differences in the definitions of the clinically relevant MACS, and heterogeneity in an individual's susceptibility to abnormal cortisol secretion from the adrenal mass. Patients with MACS present with increased risk of cardiovascular risk factors, cardiovascular events, metabolic bone disease, and mortality. Adrenalectomy improves or reverses MACS-associated comorbidities in selected patients. The current review will address diagnostic and management challenges in the care of patients with MACS, discuss data on emerging biomarkers, and suggest future directions in the field of MACS.
Collapse
Affiliation(s)
- Danae A Delivanis
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Shobana Athimulam
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Irina Bancos
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
56
|
Assessment of the Concentration of Bone Metabolism Markers: Sclerostin and FGF-23 in Children with Idiopathic Nephrotic Syndrome Treated with Glucocorticosteroids. DISEASE MARKERS 2019; 2019:9698367. [PMID: 31354894 PMCID: PMC6636590 DOI: 10.1155/2019/9698367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/18/2019] [Accepted: 03/18/2019] [Indexed: 11/17/2022]
Abstract
Recurring nature of idiopathic nephrotic syndrome (INS) and steroid dependence imply a long-term treatment with glucocorticosteroids (GCSs), which increases the risk of bone metabolism disorders. The search for new markers of that process is essential. The aims of this study were to assess the concentrations of sclerostin (Scl) and fibroblast growth factor-23 (FGF-23) in the plasma of children with INS and compare Scl and FGF-23 to existing markers of bone metabolism, mainly parathyroid hormone (PTH). The study involved 70 children, 50 with INS and 20 healthy children. Patients with INS were divided into 4 groups depending on the number of relapses and applied therapy. Significantly higher concentrations of FGF-23 and Scl were found in all patient groups with INS compared to the control group, and increase in the concentrations of examined parameters depending on the number of NS relapses was showed. In patients from the group with numerous relapses, higher concentrations of FGF-23 and Scl in the relapse phase than those in the remission phase were found. We observed positive correlation in these proteins with parathyroid hormone. Positive correlation of FGF-23 and Scl in the examined group was noted. Children having relapsing INS treated with steroids have higher levels of Scl and FGF-23 that can indicate the bone metabolism disorders. The significance of these observations requires further research.
Collapse
|
57
|
Thiele S, Hannemann A, Winzer M, Baschant U, Weidner H, Nauck M, Thakker RV, Bornhäuser M, Hofbauer LC, Rauner M. Regulation of sclerostin in glucocorticoid-induced osteoporosis (GIO) in mice and humans. Endocr Connect 2019; 8:923-934. [PMID: 31234141 PMCID: PMC6612066 DOI: 10.1530/ec-19-0104] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GC) are used for the treatment of inflammatory diseases, including various forms of arthritis. However, their use is limited, amongst others, by adverse effects on bone. The Wnt and bone formation inhibitor sclerostin was recently implicated in the pathogenesis of GC-induced osteoporosis. However, data are ambiguous. The aim of this study was to assess the regulation of sclerostin by GC using several mouse models with high GC levels and two independent cohorts of patients treated with GC. Male 24-week-old C57BL/6 and 18-week-old DBA/1 mice exposed to GC and 12-week-old mice with endogenous hypercortisolism displayed reduced bone formation as indicated by reduced levels of P1NP and increased serum sclerostin levels. The expression of sclerostin in femoral bone tissue and GC-treated bone marrow stromal cells, however, was not consistently altered. In contrast, GC dose- and time-dependently suppressed sclerostin at mRNA and protein levels in human mesenchymal stromal cells, and this effect was GC receptor dependent. In line with the human cell culture data, patients with rheumatoid arthritis (RA, n = 101) and polymyalgia rheumatica (PMR, n = 21) who were exposed to GC had lower serum levels of sclerostin than healthy age- and sex-matched controls (-40%, P < 0.01 and -26.5%, P < 0.001, respectively). In summary, sclerostin appears to be differentially regulated by GC in mice and humans as it is suppressed by GCs in humans but is not consistently altered in mice. Further studies are required to delineate the differences between GC regulation of sclerostin in mice and humans and assess whether sclerostin mediates GC-induced osteoporosis in humans.
Collapse
Affiliation(s)
- Sylvia Thiele
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Anke Hannemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Maria Winzer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Heike Weidner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine University of Oxford, Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, UK
| | - Martin Bornhäuser
- Department of Medicine I, Technische Universität Dresden, Dresden, Germany
- DFG Research Center and Cluster of Excellence for Regenerative Therapies, Technical University, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
- DFG Research Center and Cluster of Excellence for Regenerative Therapies, Technical University, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
58
|
Dalla Volta A, Formenti AM, Berruti A. Higher Risk of Fragility Fractures in Prostate Cancer Patients Treated with Combined Radium-223 and Abiraterone: Prednisone May Be the Culprit. Eur Urol 2019; 75:894-895. [DOI: 10.1016/j.eururo.2019.01.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 01/17/2019] [Indexed: 11/25/2022]
|
59
|
Adhikary S, Choudhary D, Tripathi AK, Karvande A, Ahmad N, Kothari P, Trivedi R. FGF-2 targets sclerostin in bone and myostatin in skeletal muscle to mitigate the deleterious effects of glucocorticoid on musculoskeletal degradation. Life Sci 2019; 229:261-276. [PMID: 31082400 DOI: 10.1016/j.lfs.2019.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 10/26/2022]
Abstract
AIM Myokines are associated with regulation of bone and muscle mass. However, limited information is available regarding the impact of myokines on glucocorticoid (GC) mediated adverse effects on the musculoskeletal system. This study investigates the role of myokine fibroblast growth factor-2 (FGF-2) in regulating GC-induced deleterious effects on bone and skeletal muscle. METHODS Primary osteoblast cells and C2C12 myoblast cell line were treated with FGF-2 and then exposed to dexamethasone (GC). FGF-2 mediated attenuation of the inhibitory effect of GC on osteoblast and myoblast differentiation and muscle atrophy was assessed through quantitative PCR and western blot analysis. Further, FGF-2 was administered subcutaneously to dexamethasone treated mice to collect bone and skeletal muscle tissue for in vivo analysis of bone microarchitecture, mechanical strength, histomorphometry and for histological alterations in treated tissue samples. KEY FINDINGS FGF-2 abrogated the dexamethasone induced inhibitory effect on osteoblast differentiation by modulating BMP-2 pathway and inhibiting Wnt antagonist sclerostin. Further, dexamethasone induced atrophy in C2C12 cells was mitigated by FGF-2 as evident from down regulation of atrogenes expression. FGF-2 prevented GC-induced impairment of mineral density, biomechanical strength, trabecular bone volume, cortical thickness and bone formation rate in mice. Additionally, skeletal muscle tissue from GC treated mice displayed weak myostatin immunostaining and reduced expression of atrogenes following FGF-2 treatment. SIGNIFICANCE FGF-2 mitigated GC induced effects through inhibition of sclerostin and myostatin expression in bone and muscle respectively. Taken together, this study exhibited the role of exogenous FGF-2 in sustaining osteoblastogenesis and inhibiting muscle atrophy in presence of glucocorticoid.
Collapse
Affiliation(s)
- Sulekha Adhikary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Dharmendra Choudhary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anirudha Karvande
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Naseer Ahmad
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Priyanka Kothari
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
60
|
Nadeau G, Ouimet-Grennan E, Aaron M, Drouin S, Bertout L, Shalmiev A, Beaulieu P, St-Onge P, Veilleux LN, Rauch F, Petrykey K, Laverdière C, Sinnett D, Alos N, Krajinovic M. Identification of genetic variants associated with skeletal muscle function deficit in childhood acute lymphoblastic leukemia survivors. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:33-45. [PMID: 31114288 PMCID: PMC6489684 DOI: 10.2147/pgpm.s192924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022]
Abstract
Background: Although 80% of childhood acute lymphoblastic leukemia (ALL) cases are cured with current treatment protocols, exposure to chemotherapeutics or radiation therapy during a vulnerable period of child development has been associated with a high frequency of late adverse effects (LAE). Previous observations suggest important skeletal muscle size, density and function deficits in ALL survivors. Purpose: Given that only a fraction of all patients will suffer from this particular complication, we investigated whether it could be predicted by genetic markers. Patients and methods: We analysed associations between skeletal muscle force (Fmax) and power (Pmax) and germline genetic variants from 1039 genes derived through whole-exome sequencing. Top-ranking association signals retained after correction for multiple testing were confirmed through genotyping, and further analysed through stratified analyses and multivariate models. Results: Our results show that skeletal muscle function deficit is associated with two common single nucleotide polymorphisms (SNPs) (rs2001616DUOX2, P=0.0002 (Pmax) and rs41270041ADAMTS4, P=0.02 (Fmax)) and two rare ones located in the ALOX15 gene (P=0.001 (Pmax)). These associations were further modulated by sex, body mass index and risk groups, which reflected glucocorticoid dose and radiation therapy (P≤0.02). Conclusion: Occurrence of muscle function deficit in childhood ALL is thus strongly modulated by variations in the DUOX2, ADAMTS4 and ALOX15 genes, which could lead to personalized prevention strategies in childhood ALL survivors.
Collapse
Affiliation(s)
- Geneviève Nadeau
- Department of Medicine, University of Montreal, Montreal, QC, Canada
| | | | - Michelle Aaron
- Department of Medicine, University of Montreal, Montreal, QC, Canada
| | - Simon Drouin
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Laurence Bertout
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Albert Shalmiev
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Patrick Beaulieu
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Pascal St-Onge
- Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | | | - Frank Rauch
- Division of paediatrics, Montreal Shriners Hospital for Children, Montreal, QC, Canada
| | - Kateryna Petrykey
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada
| | - Caroline Laverdière
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Hemato-Oncology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| | - Daniel Sinnett
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Hemato-Oncology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| | - Nathalie Alos
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Endocrinology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| | - Maja Krajinovic
- Department of Medicine, University of Montreal, Montreal, QC, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, QC, Canada.,Division of Hemato-Oncology, Sainte-Justine University Hospital Centre, Montreal, QC, Canada
| |
Collapse
|
61
|
Kassotis CD, Stapleton HM. Endocrine-Mediated Mechanisms of Metabolic Disruption and New Approaches to Examine the Public Health Threat. Front Endocrinol (Lausanne) 2019; 10:39. [PMID: 30792693 PMCID: PMC6374316 DOI: 10.3389/fendo.2019.00039] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/17/2019] [Indexed: 01/29/2023] Open
Abstract
Obesity and metabolic disorders are of great societal concern and generate substantial human health care costs globally. Interventions have resulted in only minimal impacts on disrupting this worsening health trend, increasing attention on putative environmental contributors. Exposure to numerous environmental contaminants have, over decades, been demonstrated to result in increased metabolic dysfunction and/or weight gain in cell and animal models, and in some cases, even in humans. There are numerous mechanisms through which environmental contaminants may contribute to metabolic dysfunction, though certain mechanisms, such as activation of the peroxisome proliferator activated receptor gamma or the retinoid x receptor, have received considerably more attention than less-studied mechanisms such as antagonism of the thyroid receptor, androgen receptor, or mitochondrial toxicity. As such, research on putative metabolic disruptors is growing rapidly, as is our understanding of molecular mechanisms underlying these effects. Concurrent with these advances, new research has evaluated current models of adipogenesis, and new models have been proposed. Only in the last several years have studies really begun to address complex mixtures of contaminants and how these mixtures may disrupt metabolic health in environmentally relevant exposure scenarios. Several studies have begun to assess environmental mixtures from various environments and study the mechanisms underlying their putative metabolic dysfunction; these studies hold real promise in highlighting crucial mechanisms driving observed organismal effects. In addition, high-throughput toxicity databases (ToxCast, etc.) may provide future benefits in prioritizing chemicals for in vivo testing, particularly once the causative molecular mechanisms promoting dysfunction are better understood and expert critiques are used to hone the databases. In this review, we will review the available literature linking metabolic disruption to endocrine-mediated molecular mechanisms, discuss the novel application of environmental mixtures and implications for in vivo metabolic health, and discuss the putative utility of applying high-throughput toxicity databases to answering complex organismal health outcome questions.
Collapse
|
62
|
Mao L, Wang M, Xi X, Dai Y, Wang N, Wang J, Xue C. Sialoglycoprotein isolated from Carassius auratus eggs promotes osteogenesis by stimulating mesenchymal stem cells to commit to osteoblast differentiation. Cell Tissue Res 2019; 376:365-376. [PMID: 30715559 DOI: 10.1007/s00441-018-2976-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022]
Abstract
In this study, we explore whether the pro-osteogenic effects of sialoglycoprotein from Carassius auratus eggs (Ca-SGP) involve mesenchymal stem cells (MSCs). Ovariectomized osteoporotic mice treated with Ca-SGP had increased bone formation and reduced bone marrow adipose tissue. As MSCs are common progenitors of osteoblasts and adipocytes, we isolated MSCs from Ca-SGP-treated mice and found that they tended to differentiate into osteoblasts over adipocytes confirmed by Alizarin red and Oil red O staining. This change was seen at the gene and protein level. To further explore the effect of Ca-SGP on MSCs, we isolated MSCs from healthy mice and treated them with Ca-SGP in vitro. We discovered that Ca-SGP promoted MSC differentiation to osteoblasts. In addition, Ca-SGP promoted osteogenesis and reduced the fat in marrow cavity of adolescent mice. For the first time, our results demonstrate that Ca-SGP promotes osteogenesis via stimulating MSCs to commit to osteoblasts. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Lei Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Meiling Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Xingjun Xi
- China National Institute of Standardization, Beijing, 100191, China
| | - Yufeng Dai
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Na Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China.
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, Shandong Province, China
| |
Collapse
|
63
|
Abstract
PURPOSE OF THIS REVIEW The goal of the review is to provide an updated understanding of the pathophysiology of glucocorticoid-induced osteoporosis and treatment recommendations. RECENT FINDINGS Glucocorticoids reduce osteoblast and osteocyte lifespan and activity and reduce the vascularity of the bone that together may explain the greater reductions in bone strength than those of bone mass. Treatments with parathyroid hormone fragments appear to reverse glucocorticoid-induced bone loss and fracture risk partially through maintaining bone vascularity and bone strength. This review identifies how glucocorticoid anti-osteogenic and vascular effects together may reduce bone strength. It also provides guidance to clinicians on rationale treatment for glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Nancy E Lane
- Center for Musculoskeletal Health, University of California at Davis Health System, 4625 Second Avenue, Suite 2006, Sacramento, CA, 95817, USA.
| |
Collapse
|
64
|
Sost Haploinsufficiency Provokes Peracute Lethal Cardiac Tamponade without Rescuing the Osteopenia in a Mouse Model of Excess Glucocorticoids. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:753-761. [PMID: 30664862 PMCID: PMC6445804 DOI: 10.1016/j.ajpath.2018.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/21/2018] [Accepted: 12/07/2018] [Indexed: 11/23/2022]
Abstract
Glucocorticoid-induced secondary osteoporosis is the most predictable side effect of this anti-inflammatory. One of the main mechanisms by which glucocorticoids achieve such deleterious outcome in bone is by antagonizing Wnt/β-catenin signaling. Sclerostin, encoded by Sost gene, is the main negative regulator of the proformative and antiresorptive role of the Wnt signaling pathway in the skeleton. It was hypothesized that the partial inactivation of sclerostin function by genetic manipulation will rescue the osteopenia induced by high endogenous glucocorticoid levels. Sost-deficient mice were crossed with an established mouse model of excess glucocorticoids, and the effects on bone mass and structure were evaluated. Sost haploinsufficiency did not rescue the low bone mass induced by high glucocorticoids. Intriguingly, the critical manifestation of Sost deficiency combined with glucocorticoid excess was sporadic, sudden, unprovoked, and nonconvulsive death. Detailed histopathologic analysis in a wide range of tissues identified peracute hemopericardium and cardiac tamponade to be the cause. These preclinical studies reveal outcomes with direct relevance to ongoing clinical trials that explore the use of antisclerostin antibodies as a treatment for osteoporosis. They particularly highlight a potential for increased cardiovascular risk and may inform improved stratification of patients who might otherwise benefit from antisclerostin antibody treatment.
Collapse
|
65
|
Zhang K, Jia Z, Yang B, Feng Q, Xu X, Yuan W, Li X, Chen X, Duan L, Wang D, Bian L. Adaptable Hydrogels Mediate Cofactor-Assisted Activation of Biomarker-Responsive Drug Delivery via Positive Feedback for Enhanced Tissue Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800875. [PMID: 30581701 PMCID: PMC6299823 DOI: 10.1002/advs.201800875] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/13/2018] [Indexed: 05/18/2023]
Abstract
The targeted and simultaneous delivery of diverse cargoes with vastly different properties by the same vehicle is highly appealing but challenging. Here, a bioactive nanocomposite hydrogel based on hyaluronic acid and self-assembled pamidronate-magnesium nanoparticles for the localized elution and on-demand simultaneous release of bioactive ions and small molecule drugs is described. The obtained nanocomposite hydrogels exhibit excellent injectability and efficient stress relaxation, thereby allowing easy injection and consequent adaptation of hydrogels to bone defects with irregular shapes. Magnesium ions released from the hydrogels promote osteogenic differentiation of the encapsulated human mesenchymal stem cells (hMSCs) and activation of alkaline phosphatase (ALP). The activated ALP subsequently catalyzes the dephosphorylation (activation) of Dex phosphate, a pro-drug of Dex, and expedites the release of Dex from hydrogels to further promote hMSC osteogenesis. This positive feedback circuit governing the activation and release of Dex significantly enhances bone regeneration at the hydrogel implantation sites. The findings suggest that these injectable nanocomposite hydrogels mediate optimized release of diverse therapeutic cargoes and effectively promote in situ bone regeneration via minimally invasive procedures.
Collapse
Affiliation(s)
- Kunyu Zhang
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatin, New TerritoriesHong KongP. R. China
| | - Zhaofeng Jia
- Shenzhen Key Laboratory of Tissue EngineeringShenzhen Laboratory of Digital Orthopeadic EngineeringShenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen518035P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen518035P. R. China
- Postgraduate institutionGuangzhou Medical UniversityGuangzhou511436P. R. China
| | - Boguang Yang
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatin, New TerritoriesHong KongP. R. China
| | - Qian Feng
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatin, New TerritoriesHong KongP. R. China
| | - Xiao Xu
- Shenzhen Key Laboratory of Tissue EngineeringShenzhen Laboratory of Digital Orthopeadic EngineeringShenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen518035P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen518035P. R. China
- Postgraduate institutionGuangzhou Medical UniversityGuangzhou511436P. R. China
| | - Weihao Yuan
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatin, New TerritoriesHong KongP. R. China
| | - Xingfu Li
- Shenzhen Key Laboratory of Tissue EngineeringShenzhen Laboratory of Digital Orthopeadic EngineeringShenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen518035P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen518035P. R. China
| | - Xiaoyu Chen
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatin, New TerritoriesHong KongP. R. China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue EngineeringShenzhen Laboratory of Digital Orthopeadic EngineeringShenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen518035P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen518035P. R. China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue EngineeringShenzhen Laboratory of Digital Orthopeadic EngineeringShenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen518035P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen518035P. R. China
- Postgraduate institutionGuangzhou Medical UniversityGuangzhou511436P. R. China
| | - Liming Bian
- Department of Biomedical EngineeringThe Chinese University of Hong KongShatin, New TerritoriesHong KongP. R. China
- Shenzhen Research InstituteThe Chinese University of Hong KongShenzhen518172P. R. China
| |
Collapse
|
66
|
Wang L, Heckmann BL, Yang X, Long H. Osteoblast autophagy in glucocorticoid-induced osteoporosis. J Cell Physiol 2018; 234:3207-3215. [PMID: 30417506 DOI: 10.1002/jcp.27335] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/10/2018] [Indexed: 02/05/2023]
Abstract
Administration of glucocorticoids is an effective strategy for treating many inflammatory and autoimmune diseases. However, glucocorticoid treatment can have adverse effects on bone, leading to glucocorticoid-induced osteoporosis (GIO), the most common form of secondary osteoporosis. Although the pathogenesis of GIO has been studied for decades, over the past ten years the autophagy machinery has been implicated as a novel mechanism. Autophagy in osteoblasts, osteocytes, and osteoclasts plays a critical role in the maintenance of bone homeostasis. Herein, we specifically discuss how osteoblast autophagy responds to glucocorticoids and its role in the development of GIO.
Collapse
Affiliation(s)
- Lufei Wang
- Oral and Craniofacial Biomedicine Program, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xianrui Yang
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Hu Long
- Department of Orthodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
67
|
Lane NE, Mohan G, Yao W, Shidara K, Lay YAE, Junjing J, Dubrovsky A, Kimmel DB. Prevalence of glucocorticoid induced osteonecrosis in the mouse is not affected by treatments that maintain bone vascularity. Bone Rep 2018; 9:181-187. [PMID: 30510976 PMCID: PMC6260230 DOI: 10.1016/j.bonr.2018.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/24/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023] Open
Abstract
Objective Determine if LLP2A-Ale or PTH (1–34) affects the prevalence of glucocorticoid-induced osteonecrosis (ON) in a mouse model. Methods Eight-week-old young adult male BALB/cJ mice were weight-randomized into Control (Con), glucocorticoid (GC)-only, or concurrent treatments with GC and LLP2A-Ale (250 μg/kg or 500 μg/kg, IV, Days 1, 14, 28) or parathyroid hormone hPTH (1–34) (40 μg/kg, 5×/week). Mice were necropsied after 45 days for qualitative evaluation of prevalent ON and quantitative evaluation of vascularity in the distal femoral epiphysis (DFE); and quantitative evaluation of bone mass, microarchitecture, and strength in the distal femoral metaphysis and lumbar vertebral body. Results The prevalence of ON was 14% in the Con group and 36% in the GC-only group (P = 0.07). The prevalence of ON did not differ among GC-only, GC + LLP2A-Ale, and GC + PTH groups. GC-only mice had significantly lower trabecular and cortical bone strength than Con, while GC + LLP2A-Ale (500 μg/kg) and GC + PTH (1–34) groups had significantly greater trabecular bone strength than the GC-only group. GC + LLP2A-Ale (250 μg/kg and 500 μg/kg) and GC + PTH had significantly higher trabecular bone volume than GC-only mice at the vertebrae, distal femoral epiphyses and distal femoral metaphyses. DFE vascularity was lower in GC-only mice than in all other groups. Conclusion Neither LLP2A-Ale nor hPTH (1–34) reduced the prevalence of GC-induced ON, compared to GC-only mice. However, GC-treated mice given LLP2A-Ale or hPTH (1–34) had better bone mass, microarchitecture, and strength in trabecular-rich regions, and higher levels of vascularity than GC-only mice.
Collapse
Affiliation(s)
- Nancy E Lane
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Geetha Mohan
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Kie Shidara
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Yu-An Evan Lay
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Jia Junjing
- Facility of Animal Science, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
| | - Alanna Dubrovsky
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Donald B Kimmel
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
68
|
Kato T, Khanh VC, Sato K, Kimura K, Yamashita T, Sugaya H, Yoshioka T, Mishima H, Ohneda O. Elevated Expression of Dkk-1 by Glucocorticoid Treatment Impairs Bone Regenerative Capacity of Adipose Tissue-Derived Mesenchymal Stem Cells. Stem Cells Dev 2018; 27:85-99. [PMID: 29084466 DOI: 10.1089/scd.2017.0199] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoids are steroid hormones used as anti-inflammatory treatments. However, this strong immunomodulation causes undesirable side effects that impair bones, such as osteoporosis. Glucocorticoid therapy is a major risk factor for developing steroid-induced osteonecrosis of the femur head (ONFH). Since ONFH is incurable, therapy with mesenchymal stem cells (MSCs) that can differentiate into osteoblasts are a first-line choice. Bone marrow-derived MSCs (BM-MSCs) are often used as a source of stem cell therapy for ONFH, but their proliferative activity is impaired after steroid treatment. Adipose tissue-derived MSCs (AT-MSCs) may be an attractive alternative source; however, it is unknown whether AT-MSCs from steroid-induced ONFH (sAT-MSCs) have the same differentiation ability as BM-MSCs or normal AT-MSCs (nAT-MSCs). In this study, we demonstrate that nAT-MSCs chronically exposed to glucocorticoids show lower alkaline phosphatase activity leading to reduced osteogenic differentiation ability. This impaired osteogenesis is mediated by high expression of Dickkopf1 (Dkk-1) that inhibits wnt/β-catenin signaling. Increased Dkk-1 also causes impaired osteogenesis along with reductions in bone regenerative capacity in sAT-MSCs. Of note, plasma Dkk-1 levels are elevated in steroid-induced ONFH patients. Collectively, our findings suggest that glucocorticoid-induced expression of Dkk-1 could be a key factor in modulating the differentiation ability of MSCs used for ONFH and other stem cell therapies.
Collapse
Affiliation(s)
- Toshiki Kato
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan .,2 School of Integrative Global Majors, University of Tsukuba , Tsukuba, Japan
| | - Vuong Cat Khanh
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Kazutoshi Sato
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Kenichi Kimura
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Toshiharu Yamashita
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Hisashi Sugaya
- 3 Department of Orthopedic Surgery, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan .,4 Division of Regenerative Medicine for Musculoskeletal System, Department of Orthopedic Surgery, Faculty of Medicine, University of Tsukuba , Tsukuba, Japan
| | - Tomokazu Yoshioka
- 3 Department of Orthopedic Surgery, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan .,4 Division of Regenerative Medicine for Musculoskeletal System, Department of Orthopedic Surgery, Faculty of Medicine, University of Tsukuba , Tsukuba, Japan
| | - Hajime Mishima
- 3 Department of Orthopedic Surgery, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| | - Osamu Ohneda
- 1 Laboratory of Regenerative Medicine and Stem Cell Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tsukuba, Japan
| |
Collapse
|
69
|
Alam I, Oakes DK, Reilly AM, Billingsley C, Sbeta S, Gerard-O'Riley RL, Acton D, Sato A, Bellido T, Econs MJ. Overexpression of WNT16 Does Not Prevent Cortical Bone Loss Due to Glucocorticoid Treatment in Mice. JBMR Plus 2018; 3:e10084. [PMID: 31044183 PMCID: PMC6478588 DOI: 10.1002/jbm4.10084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/22/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
Glucocorticoids (GC) are commonly used for the treatment of a wide variety of autoimmune, pulmonary, gastrointestinal, and malignancy conditions. One of the devastating side effects of GC use is osteoporotic fractures, particularly in the spine and hip. Bisphosphonates (BP) are the most commonly prescribed pharmacological agents for the prevention and treatment of GC-induced osteoporosis (GIO). However, GIO is marked by reduced bone formation and BP serves mainly to decrease bone resorption. The WNT signaling pathway plays a major role in bone and mineral homeostasis. Previously, we demonstrated that overexpression of WNT16 in mice led to higher bone mineral density and improved bone microarchitecture and strength. We hypothesized that WNT16 overexpression would prevent bone loss due to glucocorticoid treatment in mice. To test our hypothesis, we treated adult wild-type and WNT16-transgenic mice with vehicle and GC (prednisolone; 2.1 mg/kg body weight) via slow-release pellets for 28 days. We measured bone mass and microarchitecture by dual-energy X-ray absorptiometry (DXA) and micro-CT, and performed gene expression and serum biochemical analysis. We found that GC treatment compared with the vehicle significantly decreased femoral areal bone mineral density (aBMD), bone mineral content (BMC), and cortical bone area and thickness in both wild-type and transgenic female mice. In contrast, the trabecular bone parameters at distal femur were not significantly changed by GC treatment in male and female mice for both genotypes. Further, we observed significantly lower level of serum P1NP and a tendency of higher level of serum TRAP in wild-type and transgenic mice due to GC treatment in both sexes. Gene expression analysis showed lower mRNA levels of Wnt16, Opg, and Opg/Rankl ratio in GC-treated female mice for both genotypes compared with the sex-matched vehicle-treated mice. These data suggest that although WNT16 overexpression resulted in higher baseline bone mineral density and bone volume per trabecular volume (BV/TV) in the transgenic mice, this was insufficient to prevent bone loss in mice due to glucocorticoid treatment.
Collapse
Affiliation(s)
- Imranul Alam
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA.,Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA
| | - Dana K Oakes
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA
| | - Austin M Reilly
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA
| | - Caylin Billingsley
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA
| | - Shahed Sbeta
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA
| | | | - Dena Acton
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA
| | - Amy Sato
- Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA.,Department of Anatomy and Cell Biology Indiana University School of Medicine Indianapolis IN USA
| | - Teresita Bellido
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA.,Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA.,Department of Anatomy and Cell Biology Indiana University School of Medicine Indianapolis IN USA
| | - Michael J Econs
- Department of Medicine Indiana University School of Medicine Indianapolis IN USA.,Indiana Center for Musculoskeletal Health Indiana University School of Medicine Indianapolis IN USA.,Department of Medical and Molecular Genetics Indiana University School of Medicine Indianapolis IN USA
| |
Collapse
|
70
|
Mao L, Wang F, Li Y, Dai Y, Liu Y, Wang J, Xue C. Oil from Antarctic krill ( Euphausia superba) facilitates bone formation in dexamethasone-treated mice. Food Sci Biotechnol 2018; 28:539-545. [PMID: 30956866 DOI: 10.1007/s10068-018-0463-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/29/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022] Open
Abstract
Glucocorticoids are the leading cause of secondary osteoporosis. In the current study, the in vivo effects of Antarctic krill (Euphausia superba) oil (AKO) on dexamethasone-treated mice were investigated. Results showed that AKO significantly prevents bone loss, as evidenced by improved bone mineral density, biomechanical strength, and cancellous bone microstructure. Fluorescence double-labeling of femur showed that AKO induces new bone formation. Toluidine blue staining of marrow cavity indicated that AKO increases the number of trabecula, and decreases the generation of adipose cells. Runt-related transcription factor 2 (Runx2) and Peroxisome proliferator-activated receptor γ (PPARγ) are the switches for osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells, respectively. AKO significantly promoted the expression of Runx2 protein, and reduced PPARγ expression in bone tissue. Furthermore, AKO increased the mRNA expression of osteogenesis-related genes and decreased the expression of adipogenesis-related genes. In conclusion, AKO improved glucocorticoid-induced osteoporosis via promoting bone formation.
Collapse
Affiliation(s)
- Lei Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 Shandong Province China
| | - Fei Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 Shandong Province China
| | - Yuanyuan Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 Shandong Province China
| | - Yufeng Dai
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 Shandong Province China
| | - Yanjun Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 Shandong Province China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 Shandong Province China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003 Shandong Province China
| |
Collapse
|
71
|
Yang P, Lv S, Wang Y, Peng Y, Ye Z, Xia Z, Ding G, Cao X, Crane JL. Preservation of type H vessels and osteoblasts by enhanced preosteoclast platelet-derived growth factor type BB attenuates glucocorticoid-induced osteoporosis in growing mice. Bone 2018; 114:1-13. [PMID: 29800693 PMCID: PMC6309783 DOI: 10.1016/j.bone.2018.05.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 05/21/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022]
Abstract
Survival of chronic diseases in childhood is often achieved utilizing glucocorticoids, but comes with significant side effects, including glucocorticoid-induced osteoporosis (GIO). Knowledge of the mechanism of GIO is limited to the adult skeleton. We explored the effect of genetic loss and inhibition of cathepsin K (Ctsk) as a potential treatment target in a young GIO mouse model as genetic loss of cathepsin K results in a mild form of osteopetrosis secondary to impaired osteoclast bone resorption with maintenance of bone formation. We first characterized the temporal osteoclast and osteoblast progenitor populations in Ctsk-/- and wild type (WT) mice in the primary and secondary spongiosa, as sites representative of trabecular bone modeling and remodeling, respectively. In the primary spongiosa, Ctsk-/- mice had decreased numbers of osteoclasts at young ages (2 and 4 weeks) and increased osteoblast lineage cells at later age (8 weeks) relative to WT littermates. In the secondary spongiosa, Ctsk-/- mice had greater numbers of osteoclasts and osteoblast lineage cells relative to WT littermates. We next developed a young GIO mouse model with prednisolone 10 mg/m2/day injected intraperitoneally daily from 2 through 6 weeks of age. Overall, WT-prednisolone mice had lower bone volume per tissue volume, whereas Ctsk-/--prednisolone mice maintained a similar bone volume relative to Ctsk-/--vehicle controls. WT-prednisolone mice exhibited a decreased number of osteoclasts, tartrate-resistant acid phosphatase and platelet-derived growth factor type BB (PDGF-BB) co-positive cells, type H endothelial cells, and osteoblasts relative to WT-vehicle mice in both the primary and secondary spongiosa. Interestingly, Ctsk-/--prednisolone mice demonstrated a paradoxical response with increased numbers of all parameters in primary spongiosa and no change in secondary spongiosa. Finally, treatment with a cathepsin K inhibitor prevented WT-prednisolone decline in osteoclasts, osteoblasts, type H vessels, and bone volume. These data demonstrate that cells in the primary and secondary spongiosa respond differently to glucocorticoids and genetic manipulation. Inhibition of osteoclast resorption that preserves osteoclast coupling factors, such as through inhibition of cathepsin K, may be a potential preventive treatment strategy against GIO in the growing skeleton.
Collapse
Affiliation(s)
- Ping Yang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Obstetrics and Gynecology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi 832008, China
| | - Shan Lv
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Geriatric Endocrinology, The First Hospital Affiliated to Nanjing Medical University, Jiangsu, China
| | - Yan Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Endocrinology Department of Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, China
| | - Yi Peng
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Orthopedic Surgery, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zixing Ye
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Peking Union Medical College, Beijing, China
| | - Zhuying Xia
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute of Endocrinology and Metabolism, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guoxian Ding
- Geriatric Endocrinology, The First Hospital Affiliated to Nanjing Medical University, Jiangsu, China
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janet L Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
72
|
Kim S, Li A, Monti S, Schlezinger JJ. Tributyltin induces a transcriptional response without a brite adipocyte signature in adipocyte models. Arch Toxicol 2018; 92:2859-2874. [PMID: 30027469 DOI: 10.1101/328203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/12/2018] [Indexed: 05/24/2023]
Abstract
Tributyltin (TBT), a peroxisome proliferator-activated receptor γ (PPARγ)/retinoid X receptor (RXR) ligand and founding member of the environmental obesogen chemical class, induces adipocyte differentiation and suppresses bone formation. A growing number of environmental PPARγ ligands are being identified. However, the potential for environmental PPARγ ligands to induce adverse metabolic effects has been questioned because PPARγ is a therapeutic target in treatment of type II diabetes. We evaluated the molecular consequences of TBT exposure during bone marrow multipotent mesenchymal stromal cell (BM-MSC) differentiation in comparison to rosiglitazone, a therapeutic PPARγ ligand, and LG100268, a synthetic RXR ligand. Mouse primary BM-MSCs (female, C57BL/6J) undergoing bone differentiation were exposed to maximally efficacious and human relevant concentrations of rosiglitazone (100 nM), LG100268 (100 nM) or TBT (80 nM) for 4 days. Gene expression was assessed using microarrays, and in silico functional annotation was performed using pathway enrichment analysis approaches. Pathways related to osteogenesis were downregulated by all three ligands, while pathways related to adipogenesis were upregulated by rosiglitazone and TBT. However, pathways related to mitochondrial biogenesis and brown-in-white (brite) adipocyte differentiation were more significantly upregulated in rosiglitazone-treated than TBT-treated cells. The lack of induction of genes involved in adipocyte energy dissipation by TBT was confirmed by an independent gene expression analysis in BM-MSCs undergoing adipocyte differentiation and by analysis of a publically available 3T3 L1 data set. Furthermore, rosiglitazone, but not TBT, induced mitochondrial biogenesis and respiration. This study is the first to show that an environmental PPARγ ligand has a limited capacity to induce health-promoting activities of PPARγ.
Collapse
Affiliation(s)
- Stephanie Kim
- Department of Environmental Health, Boston University School of Public Health, 715 Albany Street, R-405, Boston, MA, 02118, USA
| | - Amy Li
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Stefano Monti
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, 715 Albany Street, R-405, Boston, MA, 02118, USA.
| |
Collapse
|
73
|
Shen G, Ren H, Shang Q, Qiu T, Yu X, Zhang Z, Huang J, Zhao W, Zhang Y, Liang D, Jiang X. Autophagy as a target for glucocorticoid-induced osteoporosis therapy. Cell Mol Life Sci 2018; 75:2683-2693. [PMID: 29427075 PMCID: PMC11105583 DOI: 10.1007/s00018-018-2776-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/25/2018] [Accepted: 02/06/2018] [Indexed: 02/07/2023]
Abstract
Autophagy takes part in regulating the eukaryotic cells function and the progression of numerous diseases, but its clinical utility has not been fully developed yet. Recently, mounting evidences highlight an important correlation between autophagy and bone homeostasis, mediated by osteoclasts, osteocytes, bone marrow mesenchymal stem cells, and osteoblasts, and autophagy plays a vital role in the pathogenesis of glucocorticoid-induced osteoporosis (GIOP). The combinations of autophagy activators/inhibitors with anti-GIOP first-line drugs or some new autophagy-based manipulators, such as regulation of B cell lymphoma 2 family proteins and caspase-dependent clearance of autophagy-related gene proteins, are likely to be the promising approaches for GIOP clinical treatments. In view of the important role of autophagy in the pathogenesis of GIOP, here we review the potential mechanisms about the impacts of autophagy in GIOP and its association with GIOP therapy.
Collapse
Affiliation(s)
- Gengyang Shen
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Hui Ren
- Department of Spinal Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ting Qiu
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jinjing Huang
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yuzhuo Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Laboratory Affiliated to National Key Discipline of Orthopaedic and Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
74
|
Abstract
PURPOSE OF REVIEW To summarize and discuss recent progress and novel signaling mechanisms relevant to bone marrow adipocyte formation and its physiological/pathophysiological implications for bone remodeling. RECENT FINDINGS Skeletal remodeling is a coordinated process entailing removal of old bone and formation of new bone. Several bone loss disorders such as osteoporosis are commonly associated with increased bone marrow adipose tissue. Experimental and clinical evidence supports that a reduction in osteoblastogenesis from mesenchymal stem cells at the expense of adipogenesis, as well as the deleterious effects of adipocyte-derived signaling, contributes to the etiology of osteoporosis as well as bone loss associated with aging, diabetes mellitus, post-menopause, and chronic drug therapy. However, this view is challenged by findings indicating that, in some contexts, bone marrow adipose tissue may have a beneficial impact on skeletal health. Further research is needed to better define the role of marrow adipocytes in bone physiology/pathophysiology and to determine the therapeutic potential of manipulating mesenchymal stem cell differentiation.
Collapse
Affiliation(s)
- Shanmugam Muruganandan
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA
| | - Christopher J Sinal
- Department of Pharmacology, Dalhousie University, 5850 College Street, Box 15000, Halifax, Nova Scotia, B3H4R2, Canada.
| |
Collapse
|
75
|
Shidara K, Mohan G, Evan Lay YA, Jepsen KJ, Yao W, Lane NE. Strain-specific differences in the development of bone loss and incidence of osteonecrosis following glucocorticoid treatment in two different mouse strains. J Orthop Translat 2018; 16:91-101. [PMID: 30723686 PMCID: PMC6350024 DOI: 10.1016/j.jot.2018.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/07/2018] [Accepted: 07/05/2018] [Indexed: 11/18/2022] Open
Abstract
Objective Glucocorticoids (GCs) are commonly prescribed as treatment for chronic inflammatory diseases. Prolonged use of GCs is a common cause of atraumatic osteonecrosis (ON) and secondary osteoporosis. Currently, there is no effective treatment for this disease; therefore, a reliable animal model would be useful to study both the pathology and novel treatment strategies for patients with the disease. The aim of this study was to establish a validated, reproducible model of GC-induced ON and bone loss in two different mouse strains (BALB/c and C57BL/6). Methods Seven-week-old male BALB/c (n = 32) and male C57BL/6 mice (n = 32) were randomised into placebo or GC groups and treated with daily 4 mg/L oral dexamethasone in drinking water for 90 days. Study outcome measures included histologic assessment of ON of the distal femur, bone mass and mechanical strength of tibia and lumbar vertebral body, osteoclast number, biochemical measure of bone formation and bone marrow fat quantitation. Results GC-induced ON lesions were observed in the distal femur in 47% of the male BALB/c mice and 25% of the male C57BL/6 mice. GC treatment decreased the trabecular bone volume and serum pro-collagen type 1N-protease (P1NP) in BALB/c mice compared with the placebo (p < 0.05) and reduced tibial bone strength in both BALB/c and C57BL/6 mice. GC-treated BALB/c mice had significantly greater marrow fat levels compared to the placebo group. Conclusion GC-induced ON was more prevalent in the male BALB/c mice compared to the male C57BL/6 mice. GC treatment significantly reduced bone mass, bone formation measured by P1NP, bone strength and increased marrow fat levels in male BALB/c mice. Therefore, the use of male BALB/c mice strain is recommended for both diagnostic and therapeutic studies for the prevention and treatment of ON and bone loss following prolonged treatment with GCs. The Translational Potential of this Article GCs are commonly used to treat patients with various chronic inflammatory diseases, and this is associated with both the development of ON and bone loss. Our study confirmed that the BALB/c mouse strain treated for 90 days with GC may be useful for developing novel treatments for ON.
Collapse
Affiliation(s)
- Kie Shidara
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, United States
| | - Geetha Mohan
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, United States
| | - Yan-An Evan Lay
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, United States
| | - Karl J. Jepsen
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, United States
| | - Nancy E. Lane
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, United States
- Corresponding author. Present address: 4625 2nd Avenue Suite 2000 Sacramento, CA, 95817, United States.
| |
Collapse
|
76
|
Huo L, Wang L, Yang Z, Li P, Geng D, Xu Y. Prednisolone induces osteoporosis-like phenotypes via focal adhesion signaling pathway in zebrafish larvae. Biol Open 2018; 7:bio.029405. [PMID: 30012552 PMCID: PMC6078353 DOI: 10.1242/bio.029405] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Patients taking glucocorticoid or glucocorticoid-like drugs for an extended period of time may develop osteoporosis, termed glucocorticoid-induced osteoporosis (GIOP). GIOP is the most common form of secondary osteoporosis, but the mechanism underlying its development is unclear. In the present study, we used prednisolone to treat zebrafish larvae to investigate GIOP. Our RNA deep-sequencing (RNA-seq) results show that prednisolone affects genes known to act in the extracellular region. Therefore the extracellular region, extracellular matrix, and collagen trimer might be involved in glucocorticoid-induced osteoporosis. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that the focal adhesion signaling pathway is the most enriched signaling pathway in terms of differentially expressed genes (DEGs). In this pathway, integrin subunit alpha 10 (itga10) and integrin subunit beta like 1 (itgbl1), genes encoding two adapter proteins, were down-regulated in the prednisolone-treated larvae. Further experiments showed that prednisolone contributes to GIOP by down-regulating itga10 and itgbl1. Summary: Glucocorticoids may contribute to osteoporosis through effects on the extracellular region, the extracellular matrix and collagen trimer, and focal adhesion signaling pathway genes (itga10 and itgbl1).
Collapse
Affiliation(s)
- Lei Huo
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shi Zi Road, Suzhou 215006, China.,Department of Orthopedics, Suzhou Science & Technology Town Hospital, 1 Lijiang Road, New District, Suzhou 215010, China
| | - Lei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shi Zi Road, Suzhou 215006, China.,Department of Orthopedics, Suzhou Science & Technology Town Hospital, 1 Lijiang Road, New District, Suzhou 215010, China
| | - Zhaoyao Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shi Zi Road, Suzhou 215006, China.,Department of Orthopedics, Suzhou Science & Technology Town Hospital, 1 Lijiang Road, New District, Suzhou 215010, China
| | - Pingyuan Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shi Zi Road, Suzhou 215006, China.,Department of Orthopedics, Suzhou Science & Technology Town Hospital, 1 Lijiang Road, New District, Suzhou 215010, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shi Zi Road, Suzhou 215006, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shi Zi Road, Suzhou 215006, China
| |
Collapse
|
77
|
Li J, Zuo B, Zhang L, Dai L, Zhang X. Osteoblast versus Adipocyte: Bone Marrow Microenvironment-Guided Epigenetic Control. CASE REPORTS IN ORTHOPEDIC RESEARCH 2018. [DOI: 10.1159/000489053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The commitment and differentiation of bone marrow mesenchymal stem cells (MSCs) is tightly controlled by the local environment ensuring lineage differentiation balance and bone homeostasis. However, pathological conditions linked with osteoporosis have changed the bone marrow microenvironment, shifting MSCs’ fate to favor adipocytes over osteoblasts, and consequently leading to decreased bone mass with marrow fat accumulation. Multiple questions related to the underlying mechanisms remain to be answered. As recent findings have confirmed the fundamental role of the epigenetic mechanism in connecting environmental signals with gene expression and stem cell differentiation, a regulatory network in the bone marrow microenvironment, epigenetic modulation, gene expression, and MSC differentiation begins to emerge. This review discusses how pathological environmental factors affect MSCs’ fate by epigenetic modulating lineage-specific genes. We conclude that manipulating local environments and/or the epigenetic regulatory machinery that target the adipocyte differentiation pathway might be a therapeutic implication of bone loss diseases such as osteoporosis.
Collapse
|
78
|
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: 16] [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
|
79
|
Dubrovsky AM, Lim MJ, Lane NE. Osteoporosis in Rheumatic Diseases: Anti-rheumatic Drugs and the Skeleton. Calcif Tissue Int 2018; 102:607-618. [PMID: 29470611 DOI: 10.1007/s00223-018-0401-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 02/01/2018] [Indexed: 12/26/2022]
Abstract
Osteoporosis in rheumatic diseases is a very well-known complication. Systemic inflammation results in both generalized and localized bone loss and erosions. Recently, increased knowledge of inflammatory process in rheumatic diseases has resulted in the development of potent inhibitors of the cytokines, the biologic DMARDs. These treatments reduce systemic inflammation and have some effect on the generalized and localized bone loss. Progression of bone erosion was slowed by TNF, IL-6 and IL-1 inhibitors, a JAK inhibitor, a CTLA4 agonist, and rituximab. Effects on bone mineral density varied between the biological DMARDs. Medications that are approved for the treatment of osteoporosis have been evaluated to prevent bone loss in rheumatic disease patients, including denosumab, cathepsin K, bisphosphonates, anti-sclerostin antibodies and parathyroid hormone (hPTH 1-34), and have some efficacy in both the prevention of systemic bone loss and reducing localized bone erosions. This article reviews the effects of biologic DMARDs on bone mass and erosions in patients with rheumatic diseases and trials of anti-osteoporotic medications in animal models and patients with rheumatic diseases.
Collapse
Affiliation(s)
- Alanna M Dubrovsky
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Mie Jin Lim
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA, 95817, USA
- Division of Rheumatology, Department of Internal Medicine, Inha University Hospital, Incheon, South Korea
| | - Nancy E Lane
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA, 95817, USA.
- Department of Internal Medicine, University of California at Davis Medical Center, 4625 2nd Avenue, Suite 2000, Sacramento, CA, 95817, USA.
| |
Collapse
|
80
|
Yang YJ, Zhu Z, Wang DT, Zhang XL, Liu YY, Lai WX, Mo YL, Li J, Liang YL, Hu ZQ, Yu YJ, Cui L. Tanshinol alleviates impaired bone formation by inhibiting adipogenesis via KLF15/PPARγ2 signaling in GIO rats. Acta Pharmacol Sin 2018; 39:633-641. [PMID: 29323335 PMCID: PMC5888681 DOI: 10.1038/aps.2017.134] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022] Open
Abstract
Glucocorticoid (GC)-induced osteoporosis (GIO) is characterized by impaired bone formation, which can be alleviated by tanshinol, an aqueous polyphenol isolated from Salvia miltiorrhiza Bunge. In this study we investigated the molecular mechanisms underlying GC-induced modulation of osteogenesis as well as the possibility of using tanshinol to interfere with GIO. Female SD rats aged 4 months were orally administered distilled water (Con), prednisone (GC, 5 mg·kg-1·d-1), GC plus tanshinol (Tan, 16 mg·kg-1·d-1) or GC plus resveratrol (Res, 5 mg·kg-1·d-1) for 14 weeks. After the rats were sacrificed, samples of bone tissues were collected. The changes in bone formation were assessed using Micro-CT, histomorphometry, and biomechanical assays. Expression of Kruppel-like factor 15 (KLF15), peroxisome proliferator-activated receptor γ 2 (PPARγ 2) and other signaling proteins in skeletal tissue was measured with Western blotting and quantitative RT-PCR. GC treatment markedly increased the expression of KLF15, PPARγ2, C/EBPα and aP2, which were related to adipogenesis, upregulated FoxO3a pathway proteins (FoxO3a and Gadd45a), and suppressed the canonical Wnt signaling (β-catenin and Axin2), which was required for osteogenesis. Thus, GC significantly decreased bone mass and bone quality. Co-treatment with Tan or Res effectively counteracted GC-impaired bone formation, suppressed GC-induced adipogenesis, and restored abnormal expression of the signaling molecules in GIO rats. We conclude that tanshinol counteracts GC-decreased bone formation by inhibiting marrow adiposity via the KLF15/PPARγ2/FoxO3a/Wnt pathway.
Collapse
Affiliation(s)
- Ya-jun Yang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Zhu Zhu
- Sino-American Cancer Research Institute, Guangdong Medical University, Dongguan 523808, China
| | - Dong-tao Wang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, China
| | - Xin-le Zhang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yu-yu Liu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Wen-xiu Lai
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yu-lin Mo
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Jin Li
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yan-long Liang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Zhuo-qing Hu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yong-jie Yu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Liao Cui
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
- Guangdong Key Laboratory for R&D of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
81
|
Soliman ME, Elmowafy E, Casettari L, Alexander C. Star-shaped poly(oligoethylene glycol) copolymer-based gels: Thermo-responsive behaviour and bioapplicability for risedronate intranasal delivery. Int J Pharm 2018; 543:224-233. [PMID: 29604369 DOI: 10.1016/j.ijpharm.2018.03.053] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/17/2018] [Accepted: 03/27/2018] [Indexed: 11/16/2022]
Abstract
The aim of this work was to obtain an intranasal delivery system with improved mechanical and mucoadhesive properties that could provide prolonged retention time for the delivery of risedronate (RS). For this, novel in situ forming gels comprising thermo-responsive star-shaped polymers, utilizing either polyethylene glycol methyl ether (PEGMA-ME 188, Mn 188) or polyethylene glycol ethyl ether (PEGMA-EE 246, Mn 246), with polyethylene glycol methyl ether (PEGMA-ME 475, Mn 475), were synthesized and characterized. RS was trapped in the selected gel-forming solutions at a concentration of 0.2% w/v. The pH, rheological properties, in vitro drug release, ex vivo permeation as well as mucoadhesion were also examined. MTT assays were conducted to verify nasal tolerability of the developed formulations. Initial in vivo studies were carried out to evaluate anti-osteoporotic activity in a glucocorticoid induced osteoporosis model in rats. The results showed successful development of thermo-sensitive formulations with favorable mechanical properties at 37 °C, which formed non-irritant, mucoadhesive porous networks, facilitating nasal RS delivery. Moreover, sustained release of RS, augmented permeability and marked anti-osteoporotic efficacy as compared to intranasal (IN) and intravenous (IV) RS solutions were realized. The combined results show that the in situ gels should have promising application as nasal drug delivery systems.
Collapse
Affiliation(s)
- Mahmoud E Soliman
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Monazzamet Elwehda Elafrikeya Street, Abbaseyya, Cairo 11566, Egypt
| | - Enas Elmowafy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Monazzamet Elwehda Elafrikeya Street, Abbaseyya, Cairo 11566, Egypt
| | - Luca Casettari
- Department of Biomolecular Sciences, School of Pharmacy, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, PU, Italy.
| | - Cameron Alexander
- School of Pharmacy, Boots Science Building, University of Nottingham, University Park, NG7 2RD Nottingham, UK
| |
Collapse
|
82
|
He H, Wang C, Tang Q, Yang F, Xu Y. Possible mechanisms of prednisolone-induced osteoporosis in zebrafish larva. Biomed Pharmacother 2018; 101:981-987. [PMID: 29635908 DOI: 10.1016/j.biopha.2018.02.082] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/06/2018] [Accepted: 02/19/2018] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is a serious clinical bone disease that results from the long-term consumption of glucocorticoids or glucocorticoid-like drugs. Although many studies have attempted to determine the mechanisms of GIOP, they are still unclear. In this study, we established a zebrafish model of glucocorticoid-like drug-induced osteoporosis by treating larvae with prednisolone. We then quantified the expression of a selection of extracellular matrix (ECM)-, osteoblast-, and osteoclast-related genes. Our results showed that at 15 days post fertilization, zebrafish larvae treated with 25 μM prednisolone are a suitable model for GIOP, not only owing to the decrease in robust bone mass but also because of significant alterations in gene expression. The quantification of the expression of ECM-, osteoblast-, and osteoclast- related genes revealed that mmp9 and mmp13 were significantly upregulated and entpd5a, acp5a, and sost were significantly downregulated. These genes may be a target for future research into GIOP. Our study thus provides new insights into GIOP.
Collapse
Affiliation(s)
- Hanliang He
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Chunqing Wang
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Qifeng Tang
- The Benq Medical Center of Suzhou, Suzhou 215000, China
| | - Fan Yang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| |
Collapse
|
83
|
Iseri K, Iyoda M, Watanabe M, Matsumoto K, Sanada D, Inoue T, Tachibana S, Shibata T. The effects of denosumab and alendronate on glucocorticoid-induced osteoporosis in patients with glomerular disease: A randomized, controlled trial. PLoS One 2018; 13:e0193846. [PMID: 29543887 PMCID: PMC5854344 DOI: 10.1371/journal.pone.0193846] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 02/16/2018] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION The clinical utility of denosumab for the treatment of glucocorticoid-induced osteoporosis (GIOP) has yet to be established. This study aimed to compare the effects of denosumab on bone mineral density (BMD) and bone turnover markers to those of alendronate in patients with GIOP. METHODS A prospective, single-center study of 32 patients (18 men; median age, 66.0 years) with glomerular disease receiving prednisolone (PSL) who were diagnosed as having GIOP and had not received bisphosphonates before was conducted. Participants were randomized to either alendronate (35 mg orally once a week) or denosumab (60 mg subcutaneously once every 6 months), and all subjects received calcitriol. The primary endpoint was the percent change in lumbar spine (LS) BMD at 12 months of treatment. RESULTS The demographic and clinical characteristics at baseline were not significantly different between the groups. Denosumab treatment markedly decreased serum levels of t-PINP, BAP, and TRACP-5b at 12 months compared to baseline (-57.4%, p<0.001; -30.9%, p<0.01; -57.7%, p<0.001, respectively). After 12 months of alendronate treatment, serum levels of t-PINP, BAP, and TRACP-5b were also significantly decreased compared to pretreatment (-38.9%, p<0.01; -16.3%, p<0.05; -43.5%, p<0.01, respectively). However, no significant differences in the changes of bone turnover markers were found between the two groups. As for the effects on BMD, denosumab treatment markedly increased LS BMD from 6 months compared to baseline, whereas no significant difference compared to pretreatment was found in the alendronate group during the study period. In the comparison of the two groups, a large increase of LS BMD was found in the denosumab treatment group compared to the alendronate treatment group at 12 months (p<0.05). CONCLUSIONS In patients with GIOP, denosumab treatment markedly suppressed bone turnover, which led to a significantly greater increase in LS BMD than with alendronate treatment. These results suggest that denosumab is a therapeutic option for the treatment of GIOP.
Collapse
Affiliation(s)
- Ken Iseri
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Nephrology Center, Makita General Hospital, Tokyo, Japan
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Makoto Watanabe
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Nephrology Center, Makita General Hospital, Tokyo, Japan
| | - Kei Matsumoto
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Daisuke Sanada
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takashi Inoue
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shohei Tachibana
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takanori Shibata
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
84
|
Adhikary S, Choudhary D, Ahmad N, Karvande A, Kumar A, Banala VT, Mishra PR, Trivedi R. Dietary flavonoid kaempferol inhibits glucocorticoid-induced bone loss by promoting osteoblast survival. Nutrition 2018; 53:64-76. [PMID: 29655780 DOI: 10.1016/j.nut.2017.12.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/13/2017] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Kaempferol, a dietary flavonoid found in fruits and vegetables, has been reported to reverse osteopenic condition in ovariectomized rats. Because kaempferol is endowed with osteogenic activity, the aim of this study was to determine whether it has a beneficial effect on glucocorticoid (GC)-induced bone loss. METHODS Adult female rats were divided into four groups as control (vehicle; distilled water), methylprednisolone (MP; 5 mg•kg•d, subcutaneously), MP + kaempferol (5 mg•kg•d, oral), and MP + human parathyroid 1-34 (30 µg/kg, 5 times/wk, subcutaneously) and treated for 4 wk. To study the antagonizing effect of kaempferol on GC-induced inhibition of fracture healing, drill-hole injury was performed on control and GC-treated rats. An oral dose of kaempferol was given for 14 d to observe the effect on callus formation at the site of injury. After treatment, bones were collected for further analysis. RESULTS GC was associated with a decreased bone mineral density and impaired bone microarchitecture parameters. Consumption of kaempferol induced bone-sparing effects in GC-induced osteopenic condition. Additionally, improved callus formation at site of drill injury in femur diaphysis was observed with kaempferol consumption in animals on GC. Consistent with the in vivo data, kaempferol elicited a higher expression of osteogenic markers in vitro and antagonized the apoptotic effect of dexamethasone on calvarial osteoblasts. CONCLUSION These results suggested that kaempferol reduced GC-induced bone loss and enhanced bone regeneration at fractured site, thus emphasizing the positive role of flavonoids on bone health.
Collapse
Affiliation(s)
- Sulekha Adhikary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Dharmendra Choudhary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Naseer Ahmad
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Anirudha Karvande
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Avinash Kumar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Venkatesh Teja Banala
- Division of Pharmaceutics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
85
|
Belaya ZE, Grebennikova TA, Melnichenko GA, Nikitin AG, Solodovnikov AG, Brovkina OI, Grigoriev AU, Rozhinskaya LY, Dedov II. Effects of endogenous hypercortisolism on bone mRNA and microRNA expression in humans. Osteoporos Int 2018; 29:211-221. [PMID: 28980049 DOI: 10.1007/s00198-017-4241-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
UNLABELLED Hypercortisolism in humans suppresses osteoblastogenesis and osteoblast function through the upregulation of Wnt-signaling antagonists (sclerostin, Dkk1) and changes in microRNAs levels (miR-125b-5p, miR-218-5p, miR-34a-5p, miR-188-3p, miR-199a-5p) which are associated with mesenchymal stem-cell commitment to adipocytes or cartilage cells over the osteoblasts. INTRODUCTION The purpose of this study was to evaluate the responses of bone to chronic glucocorticoid (GC) excess by measuring the levels of selected mRNA and microRNA (miR) in bone samples of patients with Cushing's disease (CD). METHODS Bone samples were obtained during transsphenoidal adenomectomy from the sphenoid bone (sella turcica) from 16 patients with clinically and biochemically evident CD and 10 patients with clinically non-functioning pituitary adenomas (NFPA) matched by sex, age, and body mass index. Quantitative polymerase chain reactions (qPCR) were used to examine the expression of genes (mRNA and miRs) known to be involved in bone remodeling regulation based on studies in animals and cell culture. RESULTS Hypercortisolism was associated with the downregulation of genes involved in osteoblast function and maturation (ACP5, ALPL, BGLAP, COL1A1, COL1A2, BMP2, RUNX2, TWIST1). An excess of GC caused increased expression of Wnt-signaling antagonists (Dkk1, SOST) and changes in the levels of miRs that are known to suppress osteoblastogenesis (miR-125b-5p, miR-218-5p, miR-34a-5p, miR-188-3p, miR-199a-5p) p < 0.05, q < 0.1. Interestingly, compensatory mechanisms were found in long-term hypercortisolism: upregulation of Wnt10b, LRP5, and LRP6; downregulation of SFRP4; changes in miRs involved in osteoblastogenesis (miR-210-5p, miR-135a-5p, miR-211, miR-23a-3p, miR-204-5p); and downregulation of genes associated with osteoclastogenesis. None of these changes prevented the suppression of bone formation. CONCLUSIONS An excess of endogenous GC in humans suppresses bone formation through the upregulation of Wnt-signaling antagonists and dysregulation of miRs involved in mesenchymal stem-cell commitment. Both Wnt-signaling antagonists and miRs seem to be promising targets for further research in therapeutic intervention in glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Z E Belaya
- The National Research Centre for Endocrinology, ul. Dmitria Uljanova, 11, Moscow, Russia, 117036.
| | - T A Grebennikova
- The National Research Centre for Endocrinology, ul. Dmitria Uljanova, 11, Moscow, Russia, 117036
| | - G A Melnichenko
- The National Research Centre for Endocrinology, ul. Dmitria Uljanova, 11, Moscow, Russia, 117036
| | - A G Nikitin
- Federal Research and Clinical Center FMBA of Russia, Moscow, Russia
| | | | - O I Brovkina
- Federal Research and Clinical Center FMBA of Russia, Moscow, Russia
| | - A U Grigoriev
- The National Research Centre for Endocrinology, ul. Dmitria Uljanova, 11, Moscow, Russia, 117036
| | - L Y Rozhinskaya
- The National Research Centre for Endocrinology, ul. Dmitria Uljanova, 11, Moscow, Russia, 117036
| | - I I Dedov
- The National Research Centre for Endocrinology, ul. Dmitria Uljanova, 11, Moscow, Russia, 117036
| |
Collapse
|
86
|
Wood CL, Soucek O, Wong SC, Zaman F, Farquharson C, Savendahl L, Ahmed SF. Animal models to explore the effects of glucocorticoids on skeletal growth and structure. J Endocrinol 2018; 236:R69-R91. [PMID: 29051192 DOI: 10.1530/joe-17-0361] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/17/2017] [Indexed: 12/17/2022]
Abstract
Glucocorticoids (GCs) are effective for the treatment of many chronic conditions, but their use is associated with frequent and wide-ranging adverse effects including osteoporosis and growth retardation. The mechanisms that underlie the undesirable effects of GCs on skeletal development are unclear, and there is no proven effective treatment to combat them. An in vivo model that investigates the development and progression of GC-induced changes in bone is, therefore, important and a well-characterized pre-clinical model is vital for the evaluation of new interventions. Currently, there is no established animal model to investigate GC effects on skeletal development and there are pros and cons to consider with the different protocols used to induce osteoporosis and growth retardation. This review will summarize the literature and highlight the models and techniques employed in experimental studies to date.
Collapse
Affiliation(s)
- Claire L Wood
- Division of Developmental BiologyRoslin Institute, University of Edinburgh, Edinburgh, UK
| | - Ondrej Soucek
- Department of Paediatrics2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
- Department of Women's and Children's HealthKarolinska Institutet and Pediatric Endocrinology Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Sze C Wong
- Developmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Glasgow, UK
| | - Farasat Zaman
- Department of Women's and Children's HealthKarolinska Institutet and Pediatric Endocrinology Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Colin Farquharson
- Division of Developmental BiologyRoslin Institute, University of Edinburgh, Edinburgh, UK
| | - Lars Savendahl
- Department of Women's and Children's HealthKarolinska Institutet and Pediatric Endocrinology Unit, Karolinska University Hospital, Stockholm, Sweden
| | - S Faisal Ahmed
- Developmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
87
|
Dickkopf 1 protein circulating levels as a possible biomarker of functional disability and chronic damage in patients with rheumatoid arthritis. Clin Rheumatol 2017; 37:795-801. [PMID: 29282619 DOI: 10.1007/s10067-017-3957-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/09/2017] [Accepted: 12/14/2017] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease characterized by joint destruction, deformity, lower functionality, and decrease in life expectancy. Wingless signaling pathway (Wnt) has been recently involved in bone homeostasis. Studies suggest that overexpression of the pathway inhibitors, like the Dickkopf 1 protein (DKK1), has been implicated in bone destruction. The objective of this study is to compare circulating levels of DKK1 in different groups of patients with disease activity (remission, low, moderate, high activity,) and functionality status. Three hundred seventy-nine patients with RA were evaluated between March 2015 and November 2016. Disease activity was evaluated by disease activity score 28 with C-reactive protein (DAS28CPR), simplified and clinical disease activity scores (SDAI, CDAI), routine assessment of patient index data 3 (RAPID3), functional status using Multidimensional Health Assessment Questionnaire (MD-HAQ), and the Steinbrocker functional classification. DKK1 levels were measured by ELISA. The mean age was 60.7 ± 13.9 years. Disease duration was 13.2 ± 10.9 years. Higher levels of DKK1 were not associated with disease activity by CDAI (p = 0.70), SDAI (p = 0.84), DAS28CRP (p = 0.80), or RAPID3 (p = 0.70). Interestingly higher levels of DKK1 were significantly associated to lower functional status evaluating by the Steinbrocker classification (p = 0,013), severe disability by MD-HAQ (p = 0,004), and variables associated with joint destruction like osteoporosis, higher titles of rheumatoid factor, smoking, and increased hospital admissions related to RA. Higher levels of DKK1 were found in patients with lower functional status. This association was not found in patients with greater disease activity by CDAI, SDAI, DAS28, and RAPID3. This could be explained by more structural damage; DKK1 could be used as a biomarker of joint destruction in RA.
Collapse
|
88
|
Geurtzen K, Vernet A, Freidin A, Rauner M, Hofbauer LC, Schneider JE, Brand M, Knopf F. Immune Suppressive and Bone Inhibitory Effects of Prednisolone in Growing and Regenerating Zebrafish Tissues. J Bone Miner Res 2017; 32:2476-2488. [PMID: 28771888 DOI: 10.1002/jbmr.3231] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/19/2017] [Accepted: 07/31/2017] [Indexed: 01/15/2023]
Abstract
Glucocorticoids are widely used as therapeutic agents to treat immune-mediated diseases in humans because of their anti-inflammatory and immunosuppressive effects. However, glucocorticoids have various adverse effects, in particular rapid and pronounced bone loss associated with fractures in glucocorticoid-induced osteoporosis, a common form of secondary osteoporosis. In zebrafish, which are increasingly used to study processes of bone regeneration and disease, glucocorticoids show detrimental effects on bone tissue; however, the underlying cellular mechanisms are incompletely understood. Here, we show that treatment with the glucocorticoid prednisolone impacts on the number, activity and differentiation of osteoblasts, osteoclasts, and immune cells during ontogenetic growth, homeostasis, and regeneration of zebrafish bone. Macrophage numbers are reduced in both larval and adult tissues, correlating with decreased generation of myelomonocytes and enhanced apoptosis of these cells. In contrast, osteoblasts fail to proliferate, show decreased activity, and undergo incomplete differentiation. In addition, prednisolone treatment mitigates the number and recruitment of osteoclasts to sites of bone regeneration in adult fish. In combination, these effects delay bone growth and impair bone regeneration. Our study demonstrates the many-faceted effects of glucocorticoids in non-mammalian vertebrates and helps to further establish the zebrafish as a model to study glucocorticoid-induced osteoporosis. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Karina Geurtzen
- Center for Regenerative Therapies Dresden (CRTD) and Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Aude Vernet
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Andrew Freidin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Martina Rauner
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Center for Regenerative Therapies Dresden (CRTD) and Biotechnology Center, Technische Universität Dresden, Dresden, Germany
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Jürgen E Schneider
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Michael Brand
- Center for Regenerative Therapies Dresden (CRTD) and Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Franziska Knopf
- Center for Regenerative Therapies Dresden (CRTD) and Biotechnology Center, Technische Universität Dresden, Dresden, Germany
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
89
|
Balsevich G, Petrie GN, Hill MN. Endocannabinoids: Effectors of glucocorticoid signaling. Front Neuroendocrinol 2017; 47:86-108. [PMID: 28739508 DOI: 10.1016/j.yfrne.2017.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 01/17/2023]
Abstract
For decades, there has been speculation regarding the interaction of cannabinoids with glucocorticoid systems. Given the functional redundancy between many of the physiological effects of glucocorticoids and cannabinoids, it was originally speculated that the biological mechanisms of cannabinoids were mediated by direct interactions with glucocorticoid systems. With the discovery of the endocannabinoid system, additional research demonstrated that it was actually the opposite; glucocorticoids recruit endocannabinoid signaling, and that the engagement of endocannabinoid signaling mediated many of the neurobiological and physiological effects of glucocorticoids. With the development of advances in pharmacology and genetics, significant advances in this area have been made, and it is now clear that functional interactions between these systems are critical for a wide array of physiological processes. The current review acts a comprehensive summary of the contemporary state of knowledge regarding the biological interactions between glucocorticoids and endocannabinoids, and their potential role in health and disease.
Collapse
Affiliation(s)
- Georgia Balsevich
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gavin N Petrie
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada; Departments of Cell Biology and Anatomy and Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
90
|
Prostaglandin E 2 inhibits matrix mineralization by human bone marrow stromal cell-derived osteoblasts via Epac-dependent cAMP signaling. Sci Rep 2017; 7:2243. [PMID: 28533546 PMCID: PMC5440379 DOI: 10.1038/s41598-017-02650-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023] Open
Abstract
The osteoinductive properties of prostaglandin E2 (PGE2) and its signaling pathways have led to suggestions that it may serve as a potential therapeutic strategy for bone loss. However, the prominence of PGE2 as an inducer of bone formation is attributed primarily to findings from studies using rodent models. In the current study, we investigated the effects of PGE2 on human bone marrow stromal cell (hBMSC) lineage commitment and determined its mode of action. We demonstrated that PGE2 treatment of hBMSCs significantly altered the expression profile of several genes associated with osteoblast differentiation (RUNX2 and ALP) and maturation (BGLAP and MGP). This was attributed to the activation of specific PGE2 receptors, and was associated with increases in cAMP production and sustained AKT phosphorylation. Pharmacological inhibition of exchange protein directly activated by cAMP (Epac), but not protein kinase A (PKA), recovered the mineralization functions of hBMSC-derived osteoblasts treated with PGE2 and restored AKT phosphorylation, along with the expression levels of RUNX2, ALP, BGLAP and MGP. Our findings therefore provide insights into how PGE2 influences hBMSC-mediated matrix mineralization, and should be taken into account when evaluating the role of PGE2 in human bone metabolism.
Collapse
|
91
|
Beier EE, Sheu TJ, Resseguie EA, Takahata M, Awad HA, Cory-Slechta DA, Puzas JE. Sclerostin activity plays a key role in the negative effect of glucocorticoid signaling on osteoblast function in mice. Bone Res 2017; 5:17013. [PMID: 28529816 PMCID: PMC5422922 DOI: 10.1038/boneres.2017.13] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/02/2016] [Accepted: 01/10/2017] [Indexed: 12/11/2022] Open
Abstract
Stress during prenatal development is correlated with detrimental cognitive and behavioral outcomes in offspring. However, the long-term impact of prenatal stress (PS) and disrupted glucocorticoid signaling on bone mass and strength is not understood. In contrast, the detrimental effect of lead (Pb) on skeletal health is well documented. As stress and Pb act on common biological targets via glucocorticoid signaling pathways and co-occur in the environment, this study first sought to assess the combined effect of stress and Pb on bone quality in association with alterations in glucocorticoid signaling. Bone parameters were evaluated using microCT, histomorphometry, and strength determination in 8-month-old male mouse offspring subjected to PS on gestational days 16 and 17, lifetime Pb exposure (100 p.p.m. Pb in drinking water), or to both. Pb reduced trabecular bone mass and, when combined with PS, Pb unmasked an exaggerated decrement in bone mass and tensile strength. Next, to characterize a mechanism of glucocorticoid effect on bone, prednisolone was implanted subcutaneously (controlled-release pellet, 5 mg·kg-1 per day) in 5-month-old mice that decreased osteoblastic activity and increased sclerostin and leptin levels. Furthermore, the synthetic glucocorticoid dexamethasone alters the anabolic Wnt signaling pathway. The Wnt pathway inhibitor sclerostin has several glucocorticoid response elements, and dexamethasone administration to osteoblastic cells induces sclerostin expression. Dexamethasone treatment of isolated bone marrow cells decreased bone nodule formation, whereas removal of sclerostin protected against this decrement in mineralization. Collectively, these findings suggest that bone loss associated with steroid-induced osteoporosis is a consequence of sclerostin-mediated restriction of Wnt signaling, which may mechanistically facilitate glucocorticoid toxicity in bone.
Collapse
Affiliation(s)
- Eric E Beier
- Department of Environmental Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
- Department of Environmental and Occupational Medicine, Rutgers University, Piscataway, NJ, USA
| | - Tzong-Jen Sheu
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Emily A Resseguie
- Department of Environmental Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Masahiko Takahata
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Hani A Awad
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - J Edward Puzas
- Department of Environmental Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
92
|
Fowler TW, Acevedo C, Mazur CM, Hall-Glenn F, Fields AJ, Bale HA, Ritchie RO, Lotz JC, Vail TP, Alliston T. Glucocorticoid suppression of osteocyte perilacunar remodeling is associated with subchondral bone degeneration in osteonecrosis. Sci Rep 2017; 7:44618. [PMID: 28327602 PMCID: PMC5361115 DOI: 10.1038/srep44618] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/10/2017] [Indexed: 11/09/2022] Open
Abstract
Through a process called perilacunar remodeling, bone-embedded osteocytes dynamically resorb and replace the surrounding perilacunar bone matrix to maintain mineral homeostasis. The vital canalicular networks required for osteocyte nourishment and communication, as well as the exquisitely organized bone extracellular matrix, also depend upon perilacunar remodeling. Nonetheless, many questions remain about the regulation of perilacunar remodeling and its role in skeletal disease. Here, we find that suppression of osteocyte-driven perilacunar remodeling, a fundamental cellular mechanism, plays a critical role in the glucocorticoid-induced osteonecrosis. In glucocorticoid-treated mice, we find that glucocorticoids coordinately suppress expression of several proteases required for perilacunar remodeling while causing degeneration of the osteocyte lacunocanalicular network, collagen disorganization, and matrix hypermineralization; all of which are apparent in human osteonecrotic lesions. Thus, osteocyte-mediated perilacunar remodeling maintains bone homeostasis, is dysregulated in skeletal disease, and may represent an attractive therapeutic target for the treatment of osteonecrosis.
Collapse
Affiliation(s)
- Tristan W Fowler
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Claire Acevedo
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA.,Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Courtney M Mazur
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Faith Hall-Glenn
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Aaron J Fields
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Hrishikesh A Bale
- Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Robert O Ritchie
- Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.,Department of Materials Science and Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Jeffrey C Lotz
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Thomas P Vail
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, University of California Berkeley, Berkeley, CA, USA
| |
Collapse
|
93
|
Mohan G, Lay EYA, Berka H, Ringwood L, Kot A, Chen H, Yao W, Lane NE. A Novel Hybrid Compound LLP2A-Ale Both Prevented and Rescued the Osteoporotic Phenotype in a Mouse Model of Glucocorticoid-Induced Osteoporosis. Calcif Tissue Int 2017; 100:67-79. [PMID: 27679514 PMCID: PMC5215964 DOI: 10.1007/s00223-016-0195-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
Prolonged glucocorticoid (GC) administration causes secondary osteoporosis (GIOP) and non-traumatic osteonecrosis. LLP2A-Ale is a novel bone-seeking compound that recruits mesenchymal stem cells to the bone surface, stimulates bone formation, and increases bone mass. The purpose of this study was to determine if treatment with LLP2A-Ale alone or in combination with parathyroid hormone (PTH) could prevent or treat GIOP in a mouse model. Four-month-old male Swiss-Webster mice were randomized to a prevention study with placebo, GC (day 1-28), and GC + LLP2A-Ale (IV, day 1) or a treatment study with placebo, GC (days 1-56), GC + LLP2A-Ale (IV, day 28), GC + PTH, and GC + LLP2A-Ale + PTH (days 28-56). Mice were killed on day 28 (prevention study) or on day 56 (treatment study). The study endpoints included bone mass, bone strength, serum markers of bone turnover (P1NP and CTX-I) and angiogenesis (VEGF-A), surface-based bone turnover, and blood vessel density. LLP2A-Ale prevented GC-induced bone loss and increased mechanical strength in the vertebral body (days 28 and 56) and femur (day 56). LLP2A-Ale, PTH, and LLP2A-Ale + PTH treatment significantly increased the mineralizing surface, bone formation rate, mineral apposition rate, double-labeled surface, and serum P1NP level on day 56. LLP2A-Ale and PTH treatment increased femoral blood vessel density and LLP2A-Ale increased serum VEGF-A on day 28. Therefore, LLP2A-Ale monotherapy could be a potential option to both prevent and treat GC-induced osteoporosis and bone fragility.
Collapse
Affiliation(s)
- Geetha Mohan
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Evan Yu-An Lay
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Haley Berka
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Lorna Ringwood
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Alexander Kot
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Haiyan Chen
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA
| | - Nancy E Lane
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, 4625 2nd Avenue, Suite 2006, Sacramento, CA, 95817, USA.
| |
Collapse
|
94
|
Rimando MG, Wu HH, Liu YA, Lee CW, Kuo SW, Lo YP, Tseng KF, Liu YS, Lee OKS. Glucocorticoid receptor and Histone deacetylase 6 mediate the differential effect of dexamethasone during osteogenesis of mesenchymal stromal cells (MSCs). Sci Rep 2016; 6:37371. [PMID: 27901049 PMCID: PMC5128810 DOI: 10.1038/srep37371] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/27/2016] [Indexed: 12/13/2022] Open
Abstract
Lineage commitment and differentiation of mesenchymal stromal cells (MSCs) into osteoblasts in vitro is enhanced by a potent synthetic form of glucocorticoid (GC), dexamethasone (Dex). Paradoxically, when used chronically in patients, GCs exert negative effects on bone, a phenomenon known as glucocorticoid-induced osteoporosis in clinical practice. The mechanism on how GC differentially affects bone precursor cells to become mature osteoblasts during osteogenesis remains elusive. In this study, the dose and temporal regulation of Dex on MSC differentiation into osteoblasts were investigated. We found that continuous Dex treatment led to a net reduction of the maturation potential of differentiating osteoblasts. This phenomenon correlated with a decrease in glucocorticoid receptor (GR) expression, hastened degradation, and impaired sub cellular localization. Similarly, Histone Deacetylase 6 (HDAC6) expression was found to be regulated by Dex, co-localized with GR and this GR-HDAC6 complex occupied the promoter region of the osteoblast late marker osteocalcin (OCN). Combinatorial inhibition of HDAC6 and GR enhanced OCN expression. Together, the cross-talk between the Dex effector molecule GR and the inhibitory molecule HDAC6 provided mechanistic explanation of the bimodal effect of Dex during osteogenic differentiation of MSCs. These findings may provide new directions of research to combat glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Marilyn G Rimando
- Molecular Medicine Program, Taiwan International Graduate Program, Academia Sinica and Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Hao-Hsiang Wu
- Institute of Biophotonics, National Yang-Ming University, Taipei 11221, Taiwan
| | - Yu-An Liu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Chien-Wei Lee
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 11221, Taiwan
| | - Shu-Wen Kuo
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Yin-Ping Lo
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 11221, Taiwan
| | - Kuo-Fung Tseng
- Department of Orthopaedics, Cheng-Hsin General Hospital, Taipei 11220, Taiwan
| | - Yi-Shiuan Liu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Oscar Kuang-Sheng Lee
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Taipei City Hospital, Taipei 10341, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| |
Collapse
|
95
|
Guo SC, Tao SC, Yin WJ, Qi X, Sheng JG, Zhang CQ. Exosomes from Human Synovial-Derived Mesenchymal Stem Cells Prevent Glucocorticoid-Induced Osteonecrosis of the Femoral Head in the Rat. Int J Biol Sci 2016; 12:1262-1272. [PMID: 27766040 PMCID: PMC5069447 DOI: 10.7150/ijbs.16150] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) represents a debilitating complication following glucocorticoid (GC)-based therapy. Synovial-derived mesenchymal stem cells (SMSCs) can exert protective effect in the animal model of GC-induced ONFH by inducing cell proliferation and preventing cell apoptosis. Recent studies indicate the transplanted cells exert therapeutic effects primarily via a paracrine mechanism and exosomes are an important paracrine factor that can be directly used as therapeutic agents for tissue engineering. Herein, we provided the first demonstration that the early treatment of exosomes secreted by human synovial-derived mesenchymal stem cells (SMSC-Exos) could prevent GC-induced ONFH in the rat model. Using a series of in vitro functional assays, we found that SMSC-Exos could be internalized into bone marrow derived stromal cells (BMSCs) and enhance their proliferation and have anti-apoptotic abilities. Finally, SMSC-Exos may be promising for preventing GC-induced ONFH.
Collapse
Affiliation(s)
- Shang-Chun Guo
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Shi-Cong Tao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Wen-Jing Yin
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xin Qi
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jia-Gen Sheng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Chang-Qing Zhang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China;; Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
96
|
Ahi EP. Signalling pathways in trophic skeletal development and morphogenesis: Insights from studies on teleost fish. Dev Biol 2016; 420:11-31. [PMID: 27713057 DOI: 10.1016/j.ydbio.2016.10.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/02/2016] [Accepted: 10/03/2016] [Indexed: 12/12/2022]
Abstract
During the development of the vertebrate feeding apparatus, a variety of complicated cellular and molecular processes participate in the formation and integration of individual skeletal elements. The molecular mechanisms regulating the formation of skeletal primordia and their development into specific morphological structures are tightly controlled by a set of interconnected signalling pathways. Some of these pathways, such as Bmp, Hedgehog, Notch and Wnt, are long known for their pivotal roles in craniofacial skeletogenesis. Studies addressing the functional details of their components and downstream targets, the mechanisms of their interactions with other signals as well as their potential roles in adaptive morphological divergence, are currently attracting considerable attention. An increasing number of signalling pathways that had previously been described in different biological contexts have been shown to be important in the regulation of jaw skeletal development and morphogenesis. In this review, I provide an overview of signalling pathways involved in trophic skeletogenesis emphasizing studies of the most species-rich group of vertebrates, the teleost fish, which through their evolutionary history have undergone repeated episodes of spectacular trophic diversification.
Collapse
Affiliation(s)
- Ehsan Pashay Ahi
- Institute of Zoology, University of Graz, Universitätsplatz 2, A-8010 Graz, Austria; Institute of Life and Environmental Sciences, University of Iceland, Sturlugata 7, 101 Reykjavik, Iceland.
| |
Collapse
|
97
|
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
|
98
|
Sinha S, Uchibe K, Usami Y, Pacifici M, Iwamoto M. Effectiveness and mode of action of a combination therapy for heterotopic ossification with a retinoid agonist and an anti-inflammatory agent. Bone 2016; 90:59-68. [PMID: 26891836 PMCID: PMC4970925 DOI: 10.1016/j.bone.2016.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/09/2016] [Accepted: 02/13/2016] [Indexed: 01/03/2023]
Abstract
Heterotopic ossification (HO) consists of ectopic cartilage and bone formation following severe trauma or invasive surgeries, and a genetic form of it characterizes patients with Fibrodysplasia Ossificans Progressiva (FOP). Recent mouse studies showed that HO was significantly inhibited by systemic treatment with a corticosteroid or the retinoic acid receptor γ agonist Palovarotene. Because these drugs act differently, the data raised intriguing questions including whether the drugs affected HO via similar means, whether a combination therapy would be more effective or whether the drugs may hamper each other's action. To tackle these questions, we used an effective HO mouse model involving subcutaneous implantation of Matrigel plus rhBMP2, and compared the effectiveness of prednisone, dexamathaosone, Palovarotene or combination of. Each corticosteroid and Palovarotene reduced bone formation at max doses, and a combination therapy elicited similar outcomes without obvious interference. While Palovarotene had effectively prevented the initial cartilaginous phase of HO, the steroids appeared to act more on the bony phase. In reporter assays, dexamethasone and Palovarotene induced transcriptional activity of their respective GRE or RARE constructs and did not interfere with each other's pathway. Interestingly, both drugs inhibited the activity of a reporter construct for the inflammatory mediator NF-κB, particularly in combination. In good agreement, immunohistochemical analyses showed that both drugs markedly reduced the number of mast cells and macrophages near and within the ectopic Matrigel mass and reduced also the number of progenitor cells. In sum, corticosteroids and Palovarotene appear to block HO via common and distinct mechanisms. Most importantly, they directly or indirectly inhibit the recruitment of immune and inflammatory cells present at the affected site, thus alleviating the effects of key HO instigators.
Collapse
Affiliation(s)
- Sayantani Sinha
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Kenta Uchibe
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yu Usami
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Masahiro Iwamoto
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
99
|
Chen Y, Huang L, Zhu J, Wu K. Effects of short-term glucocorticoid administration on bone mineral density, biomechanics and microstructure in rats’ femur. Hum Exp Toxicol 2016; 36:287-294. [DOI: 10.1177/0960327116649674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The effects of short-term use of oral glucocorticoid (GC) on the skeleton are not well defined. To address this gap, the influences of 7 days, 21 days of GC administration on femurs of intact rats were investigated. Forty 4-month-old female Sprague–Dawley rats were randomly divided into control group (Cont) and prednisone-treated group (Pre) and administered either distilled water or prednisone acetate at doses of 3.5 mg/kg/day for 0, 7 and 21 days, respectively. All the femurs were harvested for dual-energy X-ray absorptiometry scan, biomechanical testing and micro computed tomography scan. The whole body weight, femur bone mineral density (BMD), all three-point bending test parameters, microstructural parameters increased or improved significantly in Cont at day 21 when compared to day 0. The whole body weight, distal femur BMD, Young’s modulus, bending stiffness, density of tissue volume and trabecular thickness (Tb.Th) decreased, while structure model index and trabecular separation (Tb.Sp) increased significantly in Pre at day 21 when compared to age-matched control but had no significant differences between day 7 and day 21. Our data demonstrate that 7-day use of prednisone does not influence on rats’ femur, and 21-day use of prednisone slows in rate of whole body weight gain, decreases femur metaphysis BMD and bone stiffness which mainly due to the deteriorated bone microstructure.
Collapse
Affiliation(s)
- Y Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - L Huang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - J Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - K Wu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
100
|
Hartmann K, Koenen M, Schauer S, Wittig-Blaich S, Ahmad M, Baschant U, Tuckermann JP. Molecular Actions of Glucocorticoids in Cartilage and Bone During Health, Disease, and Steroid Therapy. Physiol Rev 2016; 96:409-47. [PMID: 26842265 DOI: 10.1152/physrev.00011.2015] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cartilage and bone are severely affected by glucocorticoids (GCs), steroid hormones that are frequently used to treat inflammatory diseases. Major complications associated with long-term steroid therapy include impairment of cartilaginous bone growth and GC-induced osteoporosis. Particularly in arthritis, GC application can increase joint and bone damage. Contrarily, endogenous GC release supports cartilage and bone integrity. In the last decade, substantial progress in the understanding of the molecular mechanisms of GC action has been gained through genome-wide binding studies of the GC receptor. These genomic approaches have revolutionized our understanding of gene regulation by ligand-induced transcription factors in general. Furthermore, specific inactivation of GC signaling and the GC receptor in bone and cartilage cells of rodent models has enabled the cell-specific effects of GCs in normal tissue homeostasis, inflammatory bone diseases, and GC-induced osteoporosis to be dissected. In this review, we summarize the current view of GC action in cartilage and bone. We further discuss future research directions in the context of new concepts for optimized steroid therapies with less detrimental effects on bone.
Collapse
Affiliation(s)
- Kerstin Hartmann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Mascha Koenen
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Schauer
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Stephanie Wittig-Blaich
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Mubashir Ahmad
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Jan P Tuckermann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|