1
|
Hu L, Chen W, Qian A, Li YP. Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res 2024; 12:39. [PMID: 38987555 PMCID: PMC11237130 DOI: 10.1038/s41413-024-00342-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/27/2024] [Accepted: 05/12/2024] [Indexed: 07/12/2024] Open
Abstract
Wnts are secreted, lipid-modified proteins that bind to different receptors on the cell surface to activate canonical or non-canonical Wnt signaling pathways, which control various biological processes throughout embryonic development and adult life. Aberrant Wnt signaling pathway underlies a wide range of human disease pathogeneses. In this review, we provide an update of Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and diseases. The Wnt proteins, receptors, activators, inhibitors, and the crosstalk of Wnt signaling pathways with other signaling pathways are summarized and discussed. We mainly review Wnt signaling functions in bone formation, homeostasis, and related diseases, and summarize mouse models carrying genetic modifications of Wnt signaling components. Moreover, the therapeutic strategies for treating bone diseases by targeting Wnt signaling, including the extracellular molecules, cytosol components, and nuclear components of Wnt signaling are reviewed. In summary, this paper reviews our current understanding of the mechanisms by which Wnt signaling regulates bone formation, homeostasis, and the efforts targeting Wnt signaling for treating bone diseases. Finally, the paper evaluates the important questions in Wnt signaling to be further explored based on the progress of new biological analytical technologies.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Airong Qian
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
2
|
An F, Song J, Chang W, Zhang J, Gao P, Wang Y, Xiao Z, Yan C. Research Progress on the Mechanism of the SFRP-Mediated Wnt Signalling Pathway Involved in Bone Metabolism in Osteoporosis. Mol Biotechnol 2024; 66:975-990. [PMID: 38194214 DOI: 10.1007/s12033-023-01018-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
Osteoporosis (OP) is a metabolic bone disease linked to an elevated fracture risk, primarily stemming from disruptions in bone metabolism. Present clinical treatments for OP merely alleviate symptoms. Hence, there exists a pressing need to identify novel targets for the clinical treatment of OP. Research indicates that the Wnt signalling pathway is modulated by serum-secreted frizzled-related protein 5 (SFRP5), potentially serving as a pivotal regulator in bone metabolism disorders. Moreover, studies confirm elevated SFRP5 expression in OP, with SFRP5 overexpression leading to the downregulation of Wnt and β-catenin proteins in the Wnt signalling pathway, as well as the expression of osteogenesis-related marker molecules such as RUNX2, ALP, and OPN. Conversely, the opposite has been reported when SFRP5 is knocked out, suggesting that SFRP5 may be a key factor involved in the regulation of bone metabolism via the Wnt signalling axis. However, the molecular mechanisms underlying the action of SFRP5-induced OP have yet to be comprehensively elucidated. This review focusses on the molecular structure and function of SFRP5 and the potential molecular mechanisms of the SFRP5-mediated Wnt signalling pathway involved in bone metabolism in OP, providing reasonable evidence for the targeted therapy of SFRP5 for the prevention and treatment of OP.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yujie Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Zhipan Xiao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
3
|
Nelson AL, Fontana G, Miclau E, Rongstad M, Murphy W, Huard J, Ehrhart N, Bahney C. Therapeutic approaches to activate the canonical Wnt pathway for bone regeneration. J Tissue Eng Regen Med 2022; 16:961-976. [PMID: 36112528 PMCID: PMC9826348 DOI: 10.1002/term.3349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/05/2022] [Accepted: 09/01/2022] [Indexed: 01/12/2023]
Abstract
Activation of the canonical Wingless-related integration site (Wnt) pathway has been shown to increase bone formation and therefore has therapeutic potential for use in orthopedic conditions. However, attempts at developing an effective strategy to achieve Wnt activation has been met with several challenges. The inherent hydrophobicity of Wnt ligands makes isolating and purifying the protein difficult. To circumvent these challenges, many have sought to target extracellular inhibitors of the Wnt pathway, such as Wnt signaling pathway inhibitors Sclerostin and Dickkopf-1, or to use small molecules, ions and proteins to increase target Wnt genes. Here, we review systemic and localized bioactive approaches to enhance bone formation or improve bone repair through antibody-based therapeutics, synthetic Wnt surrogates and scaffold doping to target canonical Wnt. We conclude with a brief review of emerging technologies, such as mRNA therapy and Clustered Regularly Interspaced Short Palindromic Repeats technology, which serve as promising approaches for future clinical translation.
Collapse
Affiliation(s)
- Anna Laura Nelson
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA
| | - GianLuca Fontana
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Elizabeth Miclau
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA
| | - Mallory Rongstad
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - William Murphy
- Department of Orthopedics and RehabilitationUniversity of Wisconsin‐MadisonMadisonWisconsinUSA,Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Johnny Huard
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Nicole Ehrhart
- School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Chelsea Bahney
- Center for Regenerative and Personalized MedicineSteadman Philippon Research Institute (SPRI)VailColoradoUSA,School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA,Department of Clinical SciencesColorado State UniversityFort CollinsColoradoUSA,Orthopaedic Trauma InstituteUniversity of California, San Francisco (UCSF)San FranciscoCaliforniaUSA
| |
Collapse
|
4
|
Maeda K, Yoshida K, Nishizawa T, Otani K, Yamashita Y, Okabe H, Hadano Y, Kayama T, Kurosaka D, Saito M. Inflammation and Bone Metabolism in Rheumatoid Arthritis: Molecular Mechanisms of Joint Destruction and Pharmacological Treatments. Int J Mol Sci 2022; 23:2871. [PMID: 35270012 PMCID: PMC8911191 DOI: 10.3390/ijms23052871] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/31/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease characterized by a variety of symptoms and pathologies often presenting with polyarthritis. The primary symptom in the initial stage is joint swelling due to synovitis. With disease progression, cartilage and bone are affected to cause joint deformities. Advanced osteoarticular destruction and deformation can cause irreversible physical disabilities. Physical disabilities not only deteriorate patients' quality of life but also have substantial medical economic effects on society. Therefore, prevention of the progression of osteoarticular destruction and deformation is an important task. Recent studies have progressively improved our understanding of the molecular mechanism by which synovitis caused by immune disorders results in activation of osteoclasts; activated osteoclasts in turn cause bone destruction and para-articular osteoporosis. In this paper, we review the mechanisms of bone metabolism under physiological and RA conditions, and we describe the effects of therapeutic intervention against RA on bone.
Collapse
Affiliation(s)
- Kazuhiro Maeda
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Ken Yoshida
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Tetsuro Nishizawa
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Kazuhiro Otani
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Yu Yamashita
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Hinako Okabe
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Yuka Hadano
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Tomohiro Kayama
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Daitaro Kurosaka
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| |
Collapse
|
5
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
6
|
Marchini M, Ashkin MR, Bellini M, Sun MMG, Workentine ML, Okuyan HM, Krawetz R, Beier F, Rolian C. A Na +/K + ATPase Pump Regulates Chondrocyte Differentiation and Bone Length Variation in Mice. Front Cell Dev Biol 2022; 9:708384. [PMID: 34970538 PMCID: PMC8712571 DOI: 10.3389/fcell.2021.708384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022] Open
Abstract
The genetic and developmental mechanisms involved in limb formation are relatively well documented, but how these mechanisms are modulated by changes in chondrocyte physiology to produce differences in limb bone length remains unclear. Here, we used high throughput RNA sequencing (RNAseq) to probe the developmental genetic basis of variation in limb bone length in Longshanks, a mouse model of experimental evolution. We find that increased tibia length in Longshanks is associated with altered expression of a few key endochondral ossification genes such as Npr3, Dlk1, Sox9, and Sfrp1, as well reduced expression of Fxyd2, a facultative subunit of the cell membrane-bound Na+/K+ ATPase pump (NKA). Next, using murine tibia and cell cultures, we show a dynamic role for NKA in chondrocyte differentiation and in bone length regulation. Specifically, we show that pharmacological inhibition of NKA disrupts chondrocyte differentiation, by upregulating expression of mesenchymal stem cell markers (Prrx1, Serpina3n), downregulation of chondrogenesis marker Sox9, and altered expression of extracellular matrix genes (e.g., collagens) associated with proliferative and hypertrophic chondrocytes. Together, Longshanks and in vitro data suggest a broader developmental and evolutionary role of NKA in regulating limb length diversity.
Collapse
Affiliation(s)
- Marta Marchini
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Mitchell R Ashkin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Melina Bellini
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Matthew Lloyd Workentine
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Hamza Malik Okuyan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Roman Krawetz
- Department of Anatomy and Cell Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Campbell Rolian
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Wagner JM, Steubing Y, Dadras M, Wallner C, Lotzien S, Huber J, Sogorski A, Sacher M, Reinkemeier F, Dittfeld S, Becerikli M, Lehnhardt M, Behr B. Wnt3a and ASCs are capable of restoring mineralization in staph aureus-infected primary murine osteoblasts. J Bone Miner Metab 2022; 40:20-28. [PMID: 34562154 DOI: 10.1007/s00774-021-01269-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 08/23/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Bone infections are one of the main reasons for impaired bone regeneration and non-union formation. In previous experimental animal studies we could already demonstrate that bone defects due to prior infections showed a markedly reduced healing capacity, which could effectively be enhanced via application of Wnt3a and Adipose-derived stromal cells (ASCs). For a more in-depth analysis, we investigated proliferation and mineralization of cultured osteoblasts infected with staph aureus and sought to investigate effects of Wnt3a and ASCs on infected osteoblasts. MATERIALS AND METHODS Primary murine osteoblasts were isolated from calvariae and infected with staph aureus. Infected osteoblasts received treatment via application of recombinant Wnt3a, ASC conditioned medium and were furthermore cocultured with ASCs. Osteoblasts were evaluated by Alamar blue assay for metabolic activity, TUNEL-assay for apoptosis, ALP and Alizarin Red staining for mineralization. In addition, immunoflourescent staining (IF) and qRT-PCR analyses were performed. RESULTS Infected osteoblasts showed a markedly reduced ability for mineralization and increased apoptosis, which could be restored to physiological levels by Wnt3a and ASC treatment. Interestingly, metabolic activity of osteoblasts seemed to be unaffected by staph aureus infection. Additional analyses of Wnt-pathway activity revealed effective enhancement of canonical Wnt-pathway activity in Wnt3a-treated osteoblasts. CONCLUSIONS In summary, we gained further osteoblast-related insights into pathomechanisms of reduced bone healing capacity upon infections.
Collapse
Affiliation(s)
| | - Yonca Steubing
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Mehran Dadras
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Christoph Wallner
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Sebastian Lotzien
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Julika Huber
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Alexander Sogorski
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Maxi Sacher
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Felix Reinkemeier
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Stephanie Dittfeld
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Mustafa Becerikli
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Marcus Lehnhardt
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| | - Björn Behr
- University Hospital BG Bergmannsheil Bochum, Bürkle-de-la-Camp Platz 1, 44789, Bochum, Germany
| |
Collapse
|
8
|
Ma Q, Wang S, Xie Z, Shen Y, Zheng B, Jiang C, Yuan P, Yu C, Li L, Zhao X, Chen J, Qin A, Fan S, Jie Z. The SFRP1 Inhibitor WAY-316606 Attenuates Osteoclastogenesis Through Dual Modulation of Canonical Wnt Signaling. J Bone Miner Res 2022; 37:152-166. [PMID: 34490916 DOI: 10.1002/jbmr.4435] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/17/2021] [Accepted: 08/29/2021] [Indexed: 11/07/2022]
Abstract
Osteoporosis, a noteworthy age-related disease induced by imbalanced osteogenesis and osteoclastogenesis, is a serious economic burden on both individuals and society. Small molecule drugs with dual effects on both bone resorption and mineralization are pressingly needed. Secreted frizzled-related protein 1 (SFRP1), a well-known extracellular repressor of canonical Wnt signaling, has been reported to regulate osteogenesis. Global SFRP1 knockout mice show significantly elevated bone mass. Although osteoclasts (OCs) express and secrete SFRP1, the role of SFRP1 produced by OCs in osteoclastogenesis and osteoporosis remains unclear. In this work, the levels of SFRP1 were found to be increased in patients with osteoporosis compared with healthy controls. Pharmacological inhibition of SFRP1 by WAY-316606 (WAY)- attenuated osteoclastogenesis and bone resorption in vitro. The expressions of OC-specific genes were suppressed by the SFRP1 inhibitor, WAY. Mechanistically, both extracellular and intracellular SFRP1 could block activation of the canonical Wnt signaling pathway, and WAY reverse the silent status of canonical Wnt through dual effects, leading to osteoclastogenesis inhibition and osteogenesis promotion. Severe osteopenia was observed in the ovariectomized (OVX) mouse model, and WAY treatment effectively improved the OVX-induced osteoporosis. In summary, this work found that SFRP1 supports OC differentiation and function, which could be attenuated by WAY through dual modulation of canonical Wnt signaling, suggesting its therapeutic potential. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Qingliang Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shiyu Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yang Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Bingjie Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chao Jiang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Liangping Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangde Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Junxin Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhiwei Jie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
9
|
Zweifler LE, Koh AJ, Daignault-Newton S, McCauley LK. Anabolic actions of PTH in murine models: two decades of insights. J Bone Miner Res 2021; 36:1979-1998. [PMID: 34101904 PMCID: PMC8596798 DOI: 10.1002/jbmr.4389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 01/19/2023]
Abstract
Parathyroid hormone (PTH) is produced by the parathyroid glands in response to low serum calcium concentrations where it targets bones, kidneys, and indirectly, intestines. The N-terminus of PTH has been investigated for decades for its ability to stimulate bone formation when administered intermittently (iPTH) and is used clinically as an effective anabolic agent for the treatment of osteoporosis. Despite great interest in iPTH and its clinical use, the mechanisms of PTH action remain complicated and not fully defined. More than 70 gene targets in more than 90 murine models have been utilized to better understand PTH anabolic actions. Because murine studies utilized wild-type mice as positive controls, a variety of variables were analyzed to better understand the optimal conditions under which iPTH functions. The greatest responses to iPTH were in male mice, with treatment starting later than 12 weeks of age, a treatment duration lasting 5-6 weeks, and a PTH dose of 30-60 μg/kg/day. This comprehensive study also evaluated these genetic models relative to the bone formative actions with a primary focus on the trabecular compartment revealing trends in critical genes and gene families relevant for PTH anabolic actions. The summation of these data revealed the gene deletions with the greatest increase in trabecular bone volume in response to iPTH. These included PTH and 1-α-hydroxylase (Pth;1α(OH)ase, 62-fold), amphiregulin (Areg, 15.8-fold), and PTH related protein (Pthrp, 10.2-fold). The deletions with the greatest inhibition of the anabolic response include deletions of: proteoglycan 4 (Prg4, -9.7-fold), low-density lipoprotein receptor-related protein 6 (Lrp6, 1.3-fold), and low-density lipoprotein receptor-related protein 5 (Lrp5, -1.0-fold). Anabolic actions of iPTH were broadly affected via multiple and diverse genes. This data provides critical insight for future research and development, as well as application to human therapeutics. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Laura E Zweifler
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Amy J Koh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | | | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA.,Department of Pathology, Medical School, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
García-Sánchez D, González-González A, García-García P, Reyes R, Pérez-Núñez MI, Riancho JA, Évora C, Rodríguez-Rey JC, Pérez-Campo FM. Effective Osteogenic Priming of Mesenchymal Stem Cells through LNA-ASOs-Mediated Sfrp1 Gene Silencing. Pharmaceutics 2021; 13:pharmaceutics13081277. [PMID: 34452242 PMCID: PMC8398380 DOI: 10.3390/pharmaceutics13081277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 01/21/2023] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has emerged as a promising approach for bone regeneration. Importantly, the beneficial effects of MSCs can be improved by modulating the expression levels of specific genes to stimulate MSC osteogenic differentiation. We have previously shown that Smurf1 silencing by using Locked Nucleic Acid-Antisense Oligonucleotides, in combination with a scaffold that sustainably releases low doses of BMP-2, was able to increase the osteogenic potential of MSCs in the presence of BMP-2 doses significantly smaller than those currently used in the clinic. This would potentially allow an important reduction in this protein in MSs-based treatments, and thus of the side effects linked to its administration. We have further improved this system by specifically targeting the Wnt pathway modulator Sfrp1. This approach not only increases MSC bone regeneration efficiency, but is also able to induce osteogenic differentiation in osteoporotic human MSCs, bypassing the need for BMP-2 induction, underscoring the regenerative potential of this system. Achieving successful osteogenesis with the sole use of LNA-ASOs, without the need of administering pro-osteogenic factors such as BMP-2, would not only reduce the cost of treatments, but would also open the possibility of targeting these LNA-ASOs specifically to MSCs in the bone marrow, allowing us to treat systemic bone loss such as that associated with osteoporosis.
Collapse
Affiliation(s)
- Daniel García-Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
| | - Alberto González-González
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
| | - Patricia García-García
- Department of Chemical Engineering and Pharmaceutical Technology, Institute of Biomedical Technologies (ITB), University of La Laguna, 38206 La Laguna, Spain; (P.G.-G.); (C.É.)
| | - Ricardo Reyes
- Department of Biochemistry, Microbiology, Cellular Biology and Genetics, Institute of Biomedical Technologies (ITB), University of La Laguna, 38200 La Laguna, Spain;
| | - María Isabel Pérez-Núñez
- Department of Traumatology, Hospital Universitario Marqués de Valdecilla, University of Cantabria, 39008 Santander, Spain;
| | - José A. Riancho
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla-IDIVAL, University of Cantabria, 39012 Santander, Spain;
| | - Carmen Évora
- Department of Chemical Engineering and Pharmaceutical Technology, Institute of Biomedical Technologies (ITB), University of La Laguna, 38206 La Laguna, Spain; (P.G.-G.); (C.É.)
| | - José Carlos Rodríguez-Rey
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
| | - Flor M. Pérez-Campo
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Cantabria-IDIVAL, 39012 Santander, Spain; (D.G.-S.); (A.G.-G.); (J.C.R.-R.)
- Correspondence: ; Tel.: +34-942-200-958
| |
Collapse
|
11
|
Zhang Q, Yu J, Chen Q, Yan H, Du H, Luo W. Regulation of pathophysiological and tissue regenerative functions of MSCs mediated via the WNT signaling pathway (Review). Mol Med Rep 2021; 24:648. [PMID: 34278470 PMCID: PMC8299209 DOI: 10.3892/mmr.2021.12287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 06/22/2021] [Indexed: 12/18/2022] Open
Abstract
Tissues have remarkable natural capabilities to regenerate for the purpose of physiological turnover and repair of damage. Adult mesenchymal stem cells (MSCs) are well known for their unique self-renewal ability, pluripotency, homing potential, paracrine effects and immunomodulation. Advanced research of the unique properties of MSCs have opened up new horizons for tissue regenerative therapies. However, certain drawbacks of the application of MSCs, such as the low survival rate of transplanted MSCs, unsatisfactory efficiency and even failure to regenerate under an unbalanced microenvironment, are concerning with regards to their wider therapeutic applications. The activity of stem cells is mainly regulated by the anatomical niche; where they are placed during their clinical and therapeutic applications. Crosstalk between various niche signals maintains MSCs in homeostasis, in which the WNT signaling pathway plays vital roles. Several external or internal stimuli have been reported to interrupt the normal bioactivity of stem cells. The irreversible tissue loss that occurs during infection at the site of tissue grafting suggests an inhibitory effect mediated by microbial infections within MSC niches. In addition, MSC-seeded tissue engineering success is difficult in various tissues, when sites of injury are under the effects of a severe infection despite the immunomodulatory properties of MSCs. In the present review, the current understanding of the way in which WNT signaling regulates MSC activity modification under physiological and pathological conditions was summarized. An effort was also made to illustrate parts of the underlying mechanism, including the inflammatory factors and their interactions with the regulatory WNT signaling pathway, aiming to promote the clinical translation of MSC-based therapy.
Collapse
Affiliation(s)
- Qingtao Zhang
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Jian Yu
- Department of Stomatology, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Qiuqiu Chen
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Honghai Yan
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Hongjiang Du
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Wenjing Luo
- Department of General Dentistry, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA 02118, USA
| |
Collapse
|
12
|
Khodabandehloo F, Taleahmad S, Aflatoonian R, Rajaei F, Zandieh Z, Nassiri-Asl M, Eslaminejad MB. Microarray analysis identification of key pathways and interaction network of differential gene expressions during osteogenic differentiation. Hum Genomics 2020; 14:43. [PMID: 33234152 PMCID: PMC7687700 DOI: 10.1186/s40246-020-00293-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/13/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Adult bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent stem cells that can differentiate into three lineages. They are suitable sources for cell-based therapy and regenerative medicine applications. This study aims to evaluate the hub genes and key pathways of differentially expressed genes (DEGs) related to osteogenesis by bioinformatics analysis in three different days. The DEGs were derived from the three different days compared with day 0. RESULTS Gene expression profiles of GSE37558 were obtained from the Gene Expression Omnibus (GEO) database. A total of 4076 DEGs were acquired on days 8, 12, and 25. Gene ontology (GO) enrichment analysis showed that the non-canonical Wnt signaling pathway and lipopolysaccharide (LPS)-mediated signaling pathway were commonly upregulated DEGs for all 3 days. KEGG pathway analysis indicated that the PI3K-Akt and focal adhesion were also commonly upregulated DEGs for all 3 days. Ten hub genes were identified by CytoHubba on days 8, 12, and 25. Then, we focused on the association of these hub genes with the Wnt pathways that had been enriched from the protein-protein interaction (PPI) by the Cytoscape plugin MCODE. CONCLUSIONS These findings suggested further insights into the roles of the PI3K/AKT and Wnt pathways and their association with osteogenesis. In addition, the stem cell microenvironment via growth factors, extracellular matrix (ECM), IGF1, IGF2, LPS, and Wnt most likely affect osteogenesis by PI3K/AKT.
Collapse
Affiliation(s)
| | - Sara Taleahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Zahra Zandieh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Nassiri-Asl
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
13
|
Jiang M, Liu R, Liu L, Kot A, Liu X, Xiao W, Jia J, Li Y, Lam KS, Yao W. Identification of osteogenic progenitor cell-targeted peptides that augment bone formation. Nat Commun 2020; 11:4278. [PMID: 32855388 PMCID: PMC7453024 DOI: 10.1038/s41467-020-17417-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 06/16/2020] [Indexed: 12/02/2022] Open
Abstract
Activation and migration of endogenous mesenchymal stromal cells (MSCs) are critical for bone regeneration. Here, we report a combinational peptide screening strategy for rapid discovery of ligands that not only bind strongly to osteogenic progenitor cells (OPCs) but also stimulate osteogenic cell Akt signaling in those OPCs. Two lead compounds are discovered, YLL3 and YLL8, both of which increase osteoprogenitor osteogenic differentiation in vitro. When given to normal or osteopenic mice, the compounds increase mineral apposition rate, bone formation, bone mass, and bone strength, as well as expedite fracture repair through stimulated endogenous osteogenesis. When covalently conjugated to alendronate, YLLs acquire an additional function resulting in a “tri-functional” compound that: (i) binds to OPCs, (ii) targets bone, and (iii) induces “pro-survival” signal. These bone-targeted, osteogenic peptides are well suited for current tissue-specific therapeutic paradigms to augment the endogenous osteogenic cells for bone regeneration and the treatment of bone loss. Activation of osteogenic cells is essential for bone regeneration. Here, the authors screen a peptide library and identify 2 compounds that promote osteogenic progenitor cell differentiation in vitro, and show that they increase bone formation and fracture repair in mice.
Collapse
Affiliation(s)
- Min Jiang
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA.,Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025, Shanghai, China
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Lixian Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Alexander Kot
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Xueping Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Junjing Jia
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Internal Medicine, The University of California at Davis Medical Center, Sacramento, CA, 95817, USA.
| |
Collapse
|
14
|
Gu H, Shi S, Xiao F, Huang Z, Xu J, Chen G, Zhou K, Lu L, Yin X. MiR-1-3p regulates the differentiation of mesenchymal stem cells to prevent osteoporosis by targeting secreted frizzled-related protein 1. Bone 2020; 137:115444. [PMID: 32447074 DOI: 10.1016/j.bone.2020.115444] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/11/2023]
Abstract
Osteoporosis (OP) is a systemic skeletal disorder with the characteristics of bone mass reduction and microarchitecture deterioration, resulting in bone fragility and increased fracture risk. A reduction in the osteoblast-differentiation of bone marrow mesenchymal stem cells (BMSCs) is considered as a basic pathogenesis of osteoporosis. miRNAs play a substantial role in the development and differentiation of BMSCs. In the present study, we found that miR-1-3p was significantly downregulated in the bones of Chinese osteoporotic patients (n = 29). Secreted frizzled-related protein 1 (SFRP1) was predicted as a target gene of miR-1-3p via the TargetScan and PicTar softwares and validated by dual-luciferase reporter assays. The findings revealed that the expression of SFRP1 was inversely correlated with miR-1-3p in osteoporotic patients. We induced mouse MSCs (mMSCs) to osteogenesis or adipogenesis and found that miR-1-3p was upregulated during osteogenesis but downregulated during adipogenesis. The overexpression of miR-1-3p stimulated osteogenesis and inhibited adipogenesis of mMSCs. In addition, ovariectomized (OVX) mice were tested and the function of miR-1-3p in vivo was explored. Immunohistochemistry and histomorphometric assays showed that in vivo inhibition of miR-1-3p increased the expression level of SFRP1 and reduced bone formation and bone mass. Furthermore, tartrate-resistant acid phosphatase (TRAP) staining indicated that the in vivo suppression of miR-1-3p promoted osteoclast activity, suggesting that miR-1-3p may influence bone mass by regulating bone resorption. It can be concluded that miR-1-3p plays a pivotal role in the pathogenesis of osteoporosis via targeting SFRP1 and may be a potential therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Si Shi
- Department of Biochemistry and Molecular Biology, School of medicine, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - Fangzhu Xiao
- Department of Orthopedics, The Fifth Hospital of Xiamen, 101 Min 'an Road, Maxiang Town, Xiang 'an District, Xiamen, Fujian Province, 361101, PR China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Guangnan Chen
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Lixia Lu
- Department of Biochemistry and Molecular Biology, School of medicine, Tongji University, 1239 Siping Road, Shanghai 200092, PR China.
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China.
| |
Collapse
|
15
|
Subclinical inflammation in the preclinical phase of rheumatoid arthritis might contribute to articular joint damage. Hum Immunol 2020; 81:726-731. [PMID: 32690328 DOI: 10.1016/j.humimm.2020.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 01/26/2023]
Abstract
The first degree relatives of rheumatoid arthritis (RA) patients have a higher risk of developing RA, which is related to the expression of autoantibodies against citrullinated proteins (ACPA). Remarkably, prior to the onset of RA, cartilage damage is already initiated, whereas ACPA autoantibodies are already expressed. Here we show that both TNF-α and IL-6 are also increased prior to the onset of RA. Furthermore, when the levels of DKK1 and Sclerostin were evaluated in first degree relatives of RA patients, we found that the serum levels of TNF- α correlate with the expression levels of both DKK1 and Sclerostin. Interestingly, when the disease is already established, the correlation of TNF- α with DKK1 is lost in RA patients, whereas the correlation of Sclerostin with both TNF- α and IL-6 is further increased. Our data suggest a subclinical inflammation in patients at high risk of developing RA, which might lead to an increase in the levels of both DKK1 and Sclerostin, contributing to joint damage in the preclinical phase of the disease linked to the expression of ACPA autoantibodies.
Collapse
|
16
|
Meszaros K, Patocs A. Glucocorticoids Influencing Wnt/β-Catenin Pathway; Multiple Sites, Heterogeneous Effects. Molecules 2020; 25:molecules25071489. [PMID: 32218328 PMCID: PMC7181001 DOI: 10.3390/molecules25071489] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoid hormones are vital; their accurate operation is a necessity at all ages and in all life situations. Glucocorticoids regulate diverse physiological processes and they use many signaling pathways to fulfill their effect. As the operation of these hormones affects many organs, the excess of glucocorticoids is actually detrimental to the whole human body. The endogenous glucocorticoid excess is a relatively rare condition, but a significant proportion of adult people uses glucocorticoid medication for the treatment of chronic illnesses, therefore they are exposed to the side effects of long-term glucocorticoid treatment. Our review summarizes the adverse effects of glucocorticoid excess affecting bones, adipose tissue, brain and skin, focusing on those effects which involve the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
| | - Attila Patocs
- Hereditary Tumours Research Group, 1089 Budapest, Hungary;
- Department of Laboratory Medicine, Semmelweis University, 1089 Budapest, Hungary
- Department of Molecular Genetics, National Institute of Oncology, 1122 Budapest, Hungary
- Correspondence: ; Tel.: +36-1-266-0926; Fax: +36-1-266-0816
| |
Collapse
|
17
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part III - Further strategies for local and systemic modulation. Clin Hemorheol Microcirc 2020; 73:439-488. [PMID: 31177207 DOI: 10.3233/ch-199104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this third in a series of reviews on adjuvant drug-assisted bone healing, further approaches aiming at influencing the healing process are discussed. Local and systemic modulation of bone metabolism is pursued with use of a number of drugs with completely different indications, which are characterized by a pleiotropic spectrum of action. These include drugs used to treat lipid disorders (HMG-CoA reductase inhibitors), hypertension (ACE inhibitors), osteoporosis (bisphosphonates), cancer (proteasome inhibitors) and others. Potential applications to enhance bone healing are discussed.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
18
|
Dai C, Jia J, Kot A, Liu X, Liu L, Jiang M, Lane NE, Wise BL, Yao W. Selective inhibition of progesterone receptor in osteochondral progenitor cells, but not in mature chondrocytes, modulated subchondral bone structures. Bone 2020; 132:115196. [PMID: 31863959 PMCID: PMC7006606 DOI: 10.1016/j.bone.2019.115196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The presence or relative proportion of progesterone nuclear receptors (PR) in different tissues may contribute to sexual dimorphism in these tissues. PR is expressed in chondrocytes, but its function is mostly unknown. We hypothesized that the PR may regulate chondrocyte metabolism and affect subchondral bone structure. METHODS We utilized genetic fate mapping and immunohistochemistry to elucidate PR expression in and effect on cartilage. To define sex-dependent and chondrocyte-specific effects of the PR on subchondral bone, we selectively deleted PR in osteochondrogenic progenitor cells marked by Prx1 (Prx1; PRcKO) and Collagen 2 (Col2; PRcKO), or in matured chondrocytes marked by aggrecan (Acan; PRcKO) and evaluated subchondral bone structure at 4 months of age. Chondrocyte aging was monitored by anti-senescence marker p16INK4a, and MMP13, one of the Senescence-Associated Secretary Phenotype (SASP) components. RESULTS Compared to wild-type (WT) mice, the female Prx1; PRcKO and the Col2; PRcKO mice had greater total subchondral bone volume and greater subchondral cortical bone thickness, with increased estimated subchondral bone stiffness and failure load in both female and male Col2; PRcKO mice. Moreover, Col2; PRcKO mice from both sexes had greater bone formation and bone strength at the femurs. In contrast, we did not observe any subchondral bone changes in Acan; PRcKO mice other than higher work-to-failure observed in the male Acan; PRcKO mice. Despite no detected difference in articular cartilage between the WT and the PR; chondrocyte conditional deletion mice, there were greater numbers of senescent chondrocytes and increased MMP13 expression, especially in the male mutant mice. CONCLUSION These findings suggest that selective inhibition of PR in osteoprogenitor cells, but not in terminally differentiated chondrocytes, induced an increased subchondral bone phenotype and high estimated subchondral bone strength, which might be associated with the development of osteoarthritis in older age.
Collapse
Affiliation(s)
- Chenlin Dai
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Junjing Jia
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Alexander Kot
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Xueping Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Lixian Liu
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Min Jiang
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Nancy E Lane
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Barton L Wise
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA; Department of Orthopaedic Surgery, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA.
| |
Collapse
|
19
|
Gennari L, Merlotti D, Falchetti A, Eller Vainicher C, Cosso R, Chiodini I. Emerging therapeutic targets for osteoporosis. Expert Opin Ther Targets 2020; 24:115-130. [PMID: 32050822 DOI: 10.1080/14728222.2020.1726889] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Osteoporosis is a chronic, skeletal disorder characterized by compromised bone strength and increased fracture risk; it affects 50% of women and 20% of men. In the past two decades, there have been substantial improvements in the pharmacotherapy of osteoporosis which have yielded potent inhibitors of bone resorption or stimulators of bone formation.Areas covered: This review discusses newly identified targets and pathways and conceptual approaches to the prevention of multiple age-related disorders. Furthermore, it summarizes existing therapeutic strategies for osteoporosis.Expert opinion: Our enhanced understanding of bone biology and the reciprocal interactions between bone and other tissues have allowed the identification of new targets that may facilitate the development of novel drugs. These drugs will hopefully achieve the uncoupling of bone formation from resorption and possibly exert a dual anabolic and antiresorptive effect on bone. Alas, limitations regarding adherence, efficacy on nonvertebral fracture prevention and the long-term adverse events still exist for currently available therapeutics. Moreover, the efficacy of most agents is limited by the tight coupling of osteoblasts and osteoclasts; hence the reduction of bone resorption invariably reduces bone formation, and vice versa. This field is very much 'a work in progress.'
Collapse
Affiliation(s)
- Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Daniela Merlotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Alberto Falchetti
- Unit for Bone Metabolism Diseases and Diabetes & Lab of Endocrine and Metabolic Research, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Cristina Eller Vainicher
- Endocrinology and Diabetology Units, Department of Medical Sciences and Community, Fondazione Ca'Granda Ospedale Maggiore Policlinico IRCCS, Milan, Italy
| | - Roberta Cosso
- EndOsMet Villa Donatello Private Hospital, Florence, Italy
| | - Iacopo Chiodini
- Unit for Bone Metabolism Diseases and Diabetes & Lab of Endocrine and Metabolic Research, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| |
Collapse
|
20
|
Feehan J, Al Saedi A, Duque G. Targeting fundamental aging mechanisms to treat osteoporosis. Expert Opin Ther Targets 2019; 23:1031-1039. [PMID: 31815563 DOI: 10.1080/14728222.2019.1702973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Osteoporotic fractures represent a growing burden of mortality, morbidity and socioeconomic cost to health-care systems worldwide. Osteoporosis is a disease uniquely associated with aging, therefore, an understanding of the physiological mechanisms underpinning its development as we age may open new avenues for therapeutic exploitation. Novel treatments, as well as refinement of the current approaches, are vital in the effort to sustain healthy, independent patients across the lifespan.Areas covered: This review covers the anabolic and catabolic pathways seen in bone maintenance, highlighting how they are changed with age, leading to osteoporosis. It will also discuss how these changes may be targeted therapeutically, in the development of new therapies, and the refinement of those already in use.Expert opinion: New effective and safe treatments for osteoporosis are still needed. Bone anabolics seem to be the most appropriate therapeutic approach to osteoporosis in older persons. Considering that bone and muscle mass synchronically decline with aging thus predisposing older persons to falls and fractures, combined therapeutic approaches to osteosarcopenia with a dual anabolic effect on muscle and bone will be a major advance in the treatment of these devastating conditions in the future.
Collapse
Affiliation(s)
- Jack Feehan
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, Australia
| | - Ahmed Al Saedi
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, Australia
| | - Gustavo Duque
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, Australia.,Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, Australia
| |
Collapse
|
21
|
Chen L, Lu Y, Li W, Ren Y, Yu M, Jiang S, Fu Y, Wang J, Peng S, Bilyk KT, Murphy KR, Zhuang X, Hune M, Zhai W, Wang W, Xu Q, Cheng CHC. The genomic basis for colonizing the freezing Southern Ocean revealed by Antarctic toothfish and Patagonian robalo genomes. Gigascience 2019; 8:5304890. [PMID: 30715292 PMCID: PMC6457430 DOI: 10.1093/gigascience/giz016] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/26/2018] [Accepted: 01/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background The Southern Ocean is the coldest ocean on Earth but a hot spot of evolution. The bottom-dwelling Eocene ancestor of Antarctic notothenioid fishes survived polar marine glaciation and underwent adaptive radiation, forming >120 species that fill all water column niches today. Genome-wide changes enabling physiological adaptations and the rapid expansion of the Antarctic notothenioids remain poorly understood. Results We sequenced and compared 2 notothenioid genomes—the cold-adapted and neutrally buoyant Antarctic toothfish Dissostichus mawsoni and the basal Patagonian robalo Eleginops maclovinus, representing the temperate ancestor. We detected >200 protein gene families that had expanded and thousands of genes that had evolved faster in the toothfish, with diverse cold-relevant functions including stress response, lipid metabolism, protein homeostasis, and freeze resistance. Besides antifreeze glycoprotein, an eggshell protein had functionally diversified to aid in cellular freezing resistance. Genomic and transcriptomic comparisons revealed proliferation of selcys–transfer RNA genes and broad transcriptional upregulation across anti-oxidative selenoproteins, signifying their prominent role in mitigating oxidative stress in the oxygen-rich Southern Ocean. We found expansion of transposable elements, temporally correlated to Antarctic notothenioid diversification. Additionally, the toothfish exhibited remarkable shifts in genetic programs towards enhanced fat cell differentiation and lipid storage, and promotion of chondrogenesis while inhibiting osteogenesis in bone development, collectively contributing to the achievement of neutral buoyancy and pelagicism. Conclusions Our study revealed a comprehensive landscape of evolutionary changes essential for Antarctic notothenioid cold adaptation and ecological expansion. The 2 genomes are valuable resources for further exploration of mechanisms underlying the spectacular notothenioid radiation in the coldest marine environment.
Collapse
Affiliation(s)
- Liangbiao Chen
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Ying Lu
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Wenhao Li
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Yandong Ren
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kuming, China
| | - Mengchao Yu
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Shouwen Jiang
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Yanxia Fu
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Jian Wang
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Sihua Peng
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Kevin T Bilyk
- Department of Animal Biology, University of Illinois at Urbana-Champaign, IL, USA
| | - Katherine R Murphy
- Department of Animal Biology, University of Illinois at Urbana-Champaign, IL, USA
| | - Xuan Zhuang
- Department of Animal Biology, University of Illinois at Urbana-Champaign, IL, USA
| | - Mathias Hune
- Fundación Ictiológica, Providencia, Santiago, Chile
| | - Wanying Zhai
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Wen Wang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kuming, China
| | - Qianghua Xu
- Internal Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) and International Research Center for Marine Biosciences (Ministry of Science and Technology) at Shanghai Ocean University, Shanghai, China
| | - Chi-Hing Christina Cheng
- Department of Animal Biology, University of Illinois at Urbana-Champaign, IL, USA.,Fundación Ictiológica, Providencia, Santiago, Chile
| |
Collapse
|
22
|
Rodrigues AM, Eusébio M, Rodrigues AB, Caetano-Lopes J, Lopes IP, Lopes A, Mendes JM, Coelho PS, Fonseca JE, Branco JC, Canhão H. Low Serum Levels of DKK2 Predict Incident Low-Impact Fracture in Older Women. JBMR Plus 2019; 3:e10179. [PMID: 31372588 PMCID: PMC6659448 DOI: 10.1002/jbm4.10179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/18/2018] [Accepted: 01/08/2019] [Indexed: 11/26/2022] Open
Abstract
There are currently no robust noninvasive markers of fragility fractures. Secreted frizzled related protein‐1 (sFRP‐1), dickkopf‐related protein 1 (DKK1) and DKK2, and sclerostin (SOST) inhibit Wnt signaling and interfere with osteoblast‐mediated bone formation. We evaluated associations of serum levels of sFRP‐1, DKK1, DKK2, and SOST with incident low‐impact fracture and BMD in 828 women aged ≥65 years from EpiDoC, a longitudinal population‐based cohort. A structured questionnaire during a baseline clinical appointment assessed prevalent fragility fractures and clinical risk factors (CRFs) for fracture. Blood was collected to measure serum levels of bone turnover markers and Wnt regulators. Lumbar spine and hip BMD were determined by DXA scanning. Follow‐up assessment was performed through a phone interview; incident fragility fracture was defined by any new self‐reported low‐impact fracture. Multivariate Cox proportional hazard models were used to analyze fracture risk adjusted for CRFs and BMD. During a mean follow‐up of 2.3 ± 1.0 years, 62 low‐impact fractures were sustained in 58 women. A low serum DKK2 level (per 1 SD decrease) was associated with a 1.5‐fold increase in fracture risk independently of BMD and CRFs. Women in the two lowest DKK2 quartiles had a fracture incidence rate of 32 per 1000 person‐years, whereas women in the two highest quartiles had 14 fragility fractures per 1000 person‐years. A high serum sFRP1 level was associated with a 1.6‐fold increase in fracture risk adjusted for CRFs, but not independently of BMD. Serum levels of SOST (r = 0.191; p = 0.0025) and DKK1(r = −0.1725; p = 0.011) were correlated with hip BMD, but not with incident fragility fracture. These results indicate that serum DKK2 and sFRP1 may predict low‐impact fracture. The low number of incident fractures recorded is a limitation and serum levels of Wnt regulators should be further studied in other populations as potential noninvasive markers of fragility fractures. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ana M Rodrigues
- CEDOC EpiDoc Unit-Epidemiology of Chronic Diseases Nova Medical School Universidade Nova de Lisboa Lisboa Portugal.,Faculdade de Medicina da Universidade de Lisboa Lisboa Portugal.,Associação EpiSaúde Évora Portugal
| | | | - Ana B Rodrigues
- Faculdade de Medicina da Universidade de Lisboa Lisboa Portugal
| | - Joana Caetano-Lopes
- Department of Orthopaedic Research Boston Children's Hospital, Boston, MA, USA; Department of Genetics Harvard Medical School Boston MA USA
| | - Inês P Lopes
- Unidade de Investigação em Reumatologia Instituto de Medicina Molecular Faculdade de Medicina Universidade de Lisboa Centro Académico de Medicina de Lisboa Lisboa Portugal
| | - Ana Lopes
- Unidade de Investigação em Reumatologia Instituto de Medicina Molecular Faculdade de Medicina Universidade de Lisboa Centro Académico de Medicina de Lisboa Lisboa Portugal
| | | | | | - João Eurico Fonseca
- Unidade de Investigação em Reumatologia Instituto de Medicina Molecular Faculdade de Medicina Universidade de Lisboa Centro Académico de Medicina de Lisboa Lisboa Portugal.,Serviço de Reumatologia e Doença Ósseas Metabólicas Hospital de Santa Maria CHLN Centro Académico de Medicina de Lisboa Lisboa Portugal
| | - Jaime C Branco
- CEDOC EpiDoc Unit-Epidemiology of Chronic Diseases Nova Medical School Universidade Nova de Lisboa Lisboa Portugal.,Centro de Estudos de Doenças Crónicas (CEDOC) da NOVA Medical School Universidade Nova de Lisboa (NMS/UNL) Lisboa Portugal.,Serviço de Reumatologia do Hospital Egas Moniz-Centro Hospitalar Lisboa Ocidental (CHLO- E.P.E.) Lisboa Portugal
| | - Helena Canhão
- CEDOC EpiDoc Unit-Epidemiology of Chronic Diseases Nova Medical School Universidade Nova de Lisboa Lisboa Portugal.,Associação EpiSaúde Évora Portugal.,Escola Nacional de Saúde Pública Universidade Nova de Lisboa Lisboa Portugal
| |
Collapse
|
23
|
Gopinathan G, Foyle D, Luan X, Diekwisch TGH. The Wnt Antagonist SFRP1: A Key Regulator of Periodontal Mineral Homeostasis. Stem Cells Dev 2019; 28:1004-1014. [PMID: 31215318 DOI: 10.1089/scd.2019.0124] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The function of mammalian periodontal tissues depends on the presence of a nonmineralized periodontal ligament (PDL) juxtaposed in between mineralized tooth anchorage tissues alveolar bone (AB) and root cementum. In the present study we have hypothesized that the Wnt antagonist secreted frizzled related protein 1 (SFRP1) is an essential regulator of periodontal tissue mineral homeostasis. Our immunoreactions and western blot data demonstrated that SFRP1 was substantially expressed higher in PDL fibroblasts than in surrounding AB progenitors and cementoblasts. SFRP1 was also detected at higher levels in PDL fibroblasts than in dental follicle (DF) cells, but the difference was less pronounced. Preferential H3K4me3 active histone mark enrichment on the SFRP1 promoter and a lack of H3K27me3 repression were most dramatic in PDL progenitors, to a lesser degree in DF cells, and not detected in AB progenitors and cementoblasts. Selective inhibition of SFRP1 using a small molecule inhibitor WAY-316606 demonstrated that SFRP1 block increased PDL cell mineralization and mineralization gene expression such as β-catenin, alkaline phosphatase, osteocalcin, collagen I, and RUNX2. The effect of SFRP1 inhibition on PDL cell mineral homeostasis was confirmed by RNA silencing. These studies also demonstrated that SFRP1 knockdown promotes PDL differentiation through histone H3K4me3-mediated activation of RUNX2 and SP7. Finally, when SFRP1 inhibition and silencing studies were performed using AB progenitors instead of PDL progenitors, there was little effect on mineralized state control and gene expression, with the exception of osteocalcin, which was dramatically upregulated upon SFRP1 silencing. Together, the results of our study document the highly specific role of the Wnt inhibitor SFRP1 in maintaining the nonmineralized state of PDL progenitors.
Collapse
Affiliation(s)
- Gokul Gopinathan
- Department of Periodontics, Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, Texas
| | - Deborah Foyle
- Department of Periodontics, Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, Texas
| | - Xianghong Luan
- Department of Periodontics, Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, Texas
| | - Thomas G H Diekwisch
- Department of Periodontics, Center for Craniofacial Research and Diagnosis, Texas A&M College of Dentistry, Dallas, Texas
| |
Collapse
|
24
|
Gu H, Wu L, Chen H, Huang Z, Xu J, Zhou K, Zhang Y, Chen J, Xia J, Yin X. Identification of differentially expressed microRNAs in the bone marrow of osteoporosis patients. Am J Transl Res 2019; 11:2940-2954. [PMID: 31217865 PMCID: PMC6556634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 02/22/2019] [Indexed: 06/09/2023]
Abstract
This study aimed to identify specific microRNAs (miRNAs) related to postmenopausal osteoporosis (OP) in human. A total of 67 conserved miRNAs, including 50 miRNAs significantly up-regulated and 17 miRNAs significantly downregulated, showed differential expression between OP group and control group. 180 hairpin structures were predicted and 199 potential novel miRNA candidates with 18 to 25 nt in length, which will greatly enrich the human miRBase. 4 miRNAs (miR-518b, miR-582-3p, miR-148a-3p and miRNA-223-3p) had upregulated expression and 4 (miR-7d-5p, miR-210-3p, miR-324-5p and miR-654-3p) showed down-regulated expression. Target genes of these miRNAs were involved in bone development, cell proliferation in bone marrow, osteoblast development, negative regulation of osteoblast differentiation, and negative regulation of osteoclast development, as well as several osteogenesis related pathways. Canonical Wnt signaling pathway was selected for verification and function analysis. The expression of Wnt1, FZD10, LRP5, DVL2 and LEF1 was down-regulated significantly, while that of SFRP1, DKK1, and CHD8 was up-regulated markedly. In conclusion, these genes play important roles in OP, which improves our understanding of pathogenesis of OP.
Collapse
Affiliation(s)
- Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Liang Wu
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Haihong Chen
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Yiming Zhang
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Jiong Chen
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Jiangni Xia
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Hospital, Fudan University Shanghai 201199, China
| |
Collapse
|
25
|
Cancer driver G-protein coupled receptor (GPCR) induced β-catenin nuclear localization: the transcriptional junction. Cancer Metastasis Rev 2019; 37:147-157. [PMID: 29222765 DOI: 10.1007/s10555-017-9711-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptors (GPCRs) comprise the main signal-transmitting components in the cell membrane. Over the past several years, biochemical and structural analyses have immensely enhanced our knowledge of GPCR involvement in health and disease states. The present review focuses on GPCRs that are cancer drivers, involved in tumor growth and development. Our aim is to highlight the involvement of stabilized β-catenin molecular machinery with a specific array of GPCRs. We discuss recent advances in understanding the molecular path leading to β-catenin nuclear localization and transcriptional activity and their implications for future cancer therapy research.
Collapse
|
26
|
Aslani S, Abhari A, Sakhinia E, Sanajou D, Rajabi H, Rahimzadeh S. Interplay between microRNAs and Wnt, transforming growth factor-β, and bone morphogenic protein signaling pathways promote osteoblastic differentiation of mesenchymal stem cells. J Cell Physiol 2018; 234:8082-8093. [PMID: 30548580 DOI: 10.1002/jcp.27582] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022]
Abstract
Osteoblasts are terminally differentiated cells with mesenchymal origins, known to possess pivotal roles in sustaining bone microstructure and homeostasis. These cells are implicated in the pathophysiology of various bone disorders, especially osteoporosis. Over the last few decades, strategies to impede bone resorption, principally by bisphosphonates, have been mainstay of treatment of osteoporosis; however, in recent years more attention has been drawn on bone-forming approaches for managing osteoporosis. MicroRNAs (miRNAs) are a broad category of noncoding short sequence RNA fragments that posttranscriptionally regulate the expression of diverse functional and structural genes in a negative manner. An accumulating body of evidence signifies that miRNAs direct mesenchymal stem cells toward osteoblast differentiation and bone formation through bone morphogenic protein, transforming growth factor-β, and Wnt signaling pathways. MiRNAs are regarded as excellent future therapeutic candidates because of their small size and ease of delivery into the cells. Considering their novel therapeutic significance, this review discusses the main miRNAs contributing to the anabolic aspects of bone formation and illustrates their interactions with corresponding signaling pathways involved in osteoblastic differentiation.
Collapse
Affiliation(s)
- Somayeh Aslani
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Abhari
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ebrahim Sakhinia
- Deparment of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Sanajou
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Rajabi
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Rahimzadeh
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Al-Barghouthi BM, Farber CR. Dissecting the Genetics of Osteoporosis using Systems Approaches. Trends Genet 2018; 35:55-67. [PMID: 30470485 DOI: 10.1016/j.tig.2018.10.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/01/2018] [Accepted: 10/22/2018] [Indexed: 02/06/2023]
Abstract
Osteoporosis is a condition characterized by low bone mineral density (BMD) and an increased risk of fracture. Traits contributing to osteoporotic fracture are highly heritable, indicating that a comprehensive understanding of bone requires a thorough understanding of the genetic basis of bone traits. Towards this goal, genome-wide association studies (GWASs) have identified over 500 loci associated with bone traits. However, few of the responsible genes have been identified, and little is known of how these genes work together to influence systems-level bone function. In this review, we describe how systems genetics approaches can be used to fill these knowledge gaps.
Collapse
Affiliation(s)
- Basel M Al-Barghouthi
- Center for Public Health Genomics, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Charles R Farber
- Center for Public Health Genomics, Departments of Public Health Sciences and Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
28
|
Ramdani G, Schall N, Kalyanaraman H, Wahwah N, Moheize S, Lee JJ, Sah RL, Pfeifer A, Casteel DE, Pilz RB. cGMP-dependent protein kinase-2 regulates bone mass and prevents diabetic bone loss. J Endocrinol 2018; 238:203-219. [PMID: 29914933 PMCID: PMC6086127 DOI: 10.1530/joe-18-0286] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 11/08/2022]
Abstract
NO/cGMP signaling is important for bone remodeling in response to mechanical and hormonal stimuli, but the downstream mediator(s) regulating skeletal homeostasis are incompletely defined. We generated transgenic mice expressing a partly-activated, mutant cGMP-dependent protein kinase type 2 (PKG2R242Q) under control of the osteoblast-specific Col1a1 promoter to characterize the role of PKG2 in post-natal bone formation. Primary osteoblasts from these mice showed a two- to three-fold increase in basal and total PKG2 activity; they proliferated faster and were resistant to apoptosis compared to cells from WT mice. Male Col1a1-Prkg2R242Q transgenic mice had increased osteoblast numbers, bone formation rates and Wnt/β-catenin-related gene expression in bone and a higher trabecular bone mass compared to their WT littermates. Streptozotocin-induced type 1 diabetes suppressed bone formation and caused rapid bone loss in WT mice, but male transgenic mice were protected from these effects. Surprisingly, we found no significant difference in bone micro-architecture or Wnt/β-catenin-related gene expression between female WT and transgenic mice; female mice of both genotypes showed higher systemic and osteoblastic NO/cGMP generation compared to their male counterparts, and a higher level of endogenous PKG2 activity may be responsible for masking effects of the PKG2R242Q transgene in females. Our data support sexual dimorphism in Wnt/β-catenin signaling and PKG2 regulation of this crucial pathway in bone homeostasis. This work establishes PKG2 as a key regulator of osteoblast proliferation and post-natal bone formation.
Collapse
Affiliation(s)
- Ghania Ramdani
- Department of MedicineUniversity of California, San Diego, La Jolla, California, USA
| | - Nadine Schall
- Department of MedicineUniversity of California, San Diego, La Jolla, California, USA
- The Institute for Pharmacology and ToxicologyUniversity of Bonn, Bonn, Germany
| | - Hema Kalyanaraman
- Department of MedicineUniversity of California, San Diego, La Jolla, California, USA
| | - Nisreen Wahwah
- Department of MedicineUniversity of California, San Diego, La Jolla, California, USA
| | - Sahar Moheize
- Department of MedicineUniversity of California, San Diego, La Jolla, California, USA
| | - Jenna J Lee
- Department of BioengineeringUniversity of California, San Diego, La Jolla, California, USA
| | - Robert L Sah
- Department of BioengineeringUniversity of California, San Diego, La Jolla, California, USA
| | - Alexander Pfeifer
- The Institute for Pharmacology and ToxicologyUniversity of Bonn, Bonn, Germany
| | - Darren E Casteel
- Department of MedicineUniversity of California, San Diego, La Jolla, California, USA
| | - Renate B Pilz
- Department of MedicineUniversity of California, San Diego, La Jolla, California, USA
| |
Collapse
|
29
|
Yu C, Xuan M, Zhang M, Yao Q, Zhang K, Zhang X, Guo J, Song L. Postnatal deletion of β-catenin in osterix-expressing cells is necessary for bone growth and intermittent PTH-induced bone gain. J Bone Miner Metab 2018; 36:560-572. [PMID: 29124436 DOI: 10.1007/s00774-017-0873-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/21/2017] [Indexed: 11/28/2022]
Abstract
wnt/β-catenin signaling has been shown to influence bone homeostasis and is important for parathyroid hormone (PTH)-induced bone gain. To further understand the role of β-catenin in the early stages of osteoblastic lineage cells for postnatal bone homeostasis and the anabolic actions of PTH on bone, we examined mice with postnatal disruption of β-catenin in osterix-expressing cells (β-catenin KO mice) by mating floxed β-catenin mice with transgenic mice expressing cre under the control of the osterix promoter suppressible by doxycycline. After withdrawal of doxycycline, β-catenin KO mice developed progressive bone loss, ectopic cartilage formation, accumulation of mesenchymal stromal cells, and bone marrow adiposity. The β-catenin-defective osteoblasts sorted by flow cytometry from β-catenin KO mice exhibited decreased EdU incorporation, increased annexin V activity, and profound alterations in gene expression including wnt target genes, osteoclast regulators, and osteoblast markers. A dramatic increase in osteoclasts was observed in both neonatal and postnatal β-catenin KO mice. Intermittent administration of PTH for 4 weeks significantly increased bone mass in control mice; however, this anabolic effect of PTH was substantially blunted in β-catenin KO mice. Our data indicate that β-catenin in osterix-expressing cells is required for postnatal osteoblast differentiation, osteoblast proliferation, and bone resorption, and is essential for the anabolic actions of PTH in bone.
Collapse
Affiliation(s)
- Caixia Yu
- Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Rd, Shanghai, 200065, China
| | - Miao Xuan
- Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Rd, Shanghai, 200065, China
| | - Mingzhu Zhang
- Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Qianqian Yao
- Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Rd, Shanghai, 200065, China
| | - Keqin Zhang
- Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Rd, Shanghai, 200065, China
| | - Xiuzhen Zhang
- Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Rd, Shanghai, 200065, China
| | - Jun Guo
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Lige Song
- Department of Endocrinology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Rd, Shanghai, 200065, China.
| |
Collapse
|
30
|
A specific haplotype in potential miRNAs binding sites of secreted frizzled-related protein 1 (SFRP1) is associated with BMD variation in osteoporosis. Gene 2018; 677:132-141. [PMID: 30055306 DOI: 10.1016/j.gene.2018.07.061] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/06/2018] [Accepted: 07/24/2018] [Indexed: 11/22/2022]
Abstract
PURPOSE Osteoporosis is an important multifactorial disease which is largely influenced by Wnt signaling pathway. Considering regulatory single nucleotide polymorphisms in Wnt signaling pathway may pave the road of understanding the genetic basis of predisposition to osteoporosis. The aim of this study was to determine the possible association between variants of SFRP1 and WNT5b, and osteoporosis incidence risk. METHODS The study population comprised 186 osteoporotic patients and 118 normal subjects from Amirkola Health and Ageing Project. rs1127379 (c.1406A>G) and rs3242 (c.3132C>T) variants in 3'UTR of SFRP1 gene, and rs3803164 (c.236C>T) in 3'UTR and rs735890 (c.622-536A>G) in intron 4 of WNT5b gene were genotyped using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Regression analyses were used to calculate the association of genotype frequencies with bone mineral density (BMD) and bone mineral content (BMC) values of participants. Bioinformatics algorithms were used to detect the effect of each SNP on the secondary structure of mRNA, and predict putative 3'UTR microRNA target sites and splicing sites changes by related SNPs. RESULTS WNT5b rs735890 was associated with lumbar spine BMD, BMC, and femoral neck BMC (P = 0.035, P = 0.007, and P = 0.038, respectively). WNT5b rs3803164, and SFRP1 rs3242 were significantly associated with lumbar spine BMD (P = 0.028 and P = 0.030, respectively). SFRP1 rs1127379 was associated with lumbar spine BMD in the male gender. Haplotype analysis showed a significant association of SFRP1 c.[1406A; 3132C] haplotype with lumbar spine BMD, and BMC (P = 0.019 and P = 0.030, respectively), and SFRP1 c.[1406G; 3132C] haplotype with lumbar spine BMC (P = 0.045). In silico analyses revealed that the G allele of SFRP1 rs1127379, and WNT5b rs3803164 appear as more possible target sites for many miRNAs. CONCLUSIONS This study is the first evidence of the association of WNT5b rs735890, and c.[1406A; 3132C] and c.[1406G; 3132C] haplotypes of SFRP1 with BMD variation in osteoporosis, probably by altering microRNA target sites, in elderly persons.
Collapse
|
31
|
Zhong ZA, Kot A, Lay YAE, Zhang H, Jia J, Lane NE, Yao W. Sex-Dependent, Osteoblast Stage-Specific Effects of Progesterone Receptor on Bone Acquisition. J Bone Miner Res 2017; 32:1841-1852. [PMID: 28569405 PMCID: PMC5611815 DOI: 10.1002/jbmr.3186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/22/2017] [Accepted: 05/27/2017] [Indexed: 12/12/2022]
Abstract
The role of the progesterone receptor (PR) in the regulation of sexual dimorphism in bone has yet to be determined. Here we utilized genetic fate mapping and Western blotting to demonstrate age-dependent PR expression in the mouse femoral metaphysis and diaphysis. To define sex-dependent and osteoblast stage-specific effects of PR on bone acquisition, we selectively deleted PR at different stages of osteoblast differentiation. We found that when Prx1-Cre mice were crossed with PR floxed mice to generate a mesenchymal stem cell (MSC) conditional KO model (Prx1; PRcKO), the mutant mice developed greater trabecular bone volume with higher mineral apposition rate and bone formation. This may be explained by increased number of MSCs and greater osteogenic potential, particularly in males. Age-related trabecular bone loss was similar between the Prx1; PRcKO mice and their WT littermates in both sexes. Hormone deficiency during the period of rapid bone growth induced rapid trabecular bone loss in both the WT and the Prx1; PRcKO mice in both sexes. No differences in trabecular bone mass was observed when PR was deleted in mature osteoblasts using osteocalcin-Cre (Bglap-Cre). Also, there were no differences in cortical bone mass in all three PRcKO mice. In conclusion, PR inactivation in early osteoprogenitor cells but not in mature osteoblasts influenced trabecular bone accrual in a sex-dependent manner. PR deletion in osteoblast lineage cells did not affect cortical bone mass. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Zhendong A. Zhong
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
- Center for Cancer and Cell Biology, Program in Skeletal Disease and Tumor Microenvironment, Van Andel Research Institute, Grand Rapids MI 49503, USA
| | - Alexander Kot
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Yu-An E. Lay
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Hongliang Zhang
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
- Department of Emergency Medicine, Center for Rare Diseases, Second Xiangya Hospital of the Central-South University, Hunan, Changsha, China
| | - Junjing Jia
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Nancy E. Lane
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| |
Collapse
|
32
|
Zhang H, Kot A, Lay YAE, Fierro FA, Chen H, Lane NE, Yao W. Acceleration of Fracture Healing by Overexpression of Basic Fibroblast Growth Factor in the Mesenchymal Stromal Cells. Stem Cells Transl Med 2017; 6:1880-1893. [PMID: 28792122 PMCID: PMC6430058 DOI: 10.1002/sctm.17-0039] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/20/2017] [Indexed: 12/29/2022] Open
Abstract
In this study, we engineered mesenchymal stem cells (MSCs) to over‐express basic fibroblast growth factor (bFGF) and evaluated its effects on fracture healing. Adipose‐derived mouse MSCs were transduced to express bFGF and green fluorescence protein (ADSCbFGF‐GFP). Closed‐femoral fractures were performed with osterix‐mCherry reporter mice of both sexes. The mice received 3 × 105 ADSCs transfected with control vector or bFGF via intramuscular injection within or around the fracture sites. Mice were euthanized at days 7, 14, and 35 to monitor MSC engraftment, osteogenic differentiation, callus formation, and bone strength. Compared to ADSC culture alone, ADSCbFGF increased bFGF expression and higher levels of bFGF and vascular endothelial growth factor (VEGF) in the culture supernatant for up to 14 days. ADSCbFGF treatment increased GFP‐labeled MSCs at the fracture gaps and these cells were incorporated into the newly formed callus. quantitative reverse transcription polymerase chain reaction (qRT‐PCR) from the callus revealed a 2‐ to 12‐fold increase in the expression of genes associated with nervous system regeneration, angiogenesis, and matrix formation. Compared to the control, ADSCbFGF treatment increased VEGF expression at the periosteal region of the callus, remodeling of collagen into mineralized callus and bone strength. In summary, MSCbFGF accelerated fracture healing by increasing the production of growth factors that stimulated angiogenesis and differentiation of MSCs to osteoblasts that formed new bone and accelerated fracture repair. This novel treatment may reduce the time required for fracture healing. Stem Cells Translational Medicine2017;6:1880–1893
Collapse
Affiliation(s)
- Hongliang Zhang
- Center for Musculoskeletal Health, Department of Internal Medicine, Sacramento, California, USA.,Department of Emergency Medicine, Center for Difficult Diagnoses and Rare Diseases, Second Xiangya Hospital of the Central-South University, Hunan, Changsha, People's Republic of China
| | - Alexander Kot
- Center for Musculoskeletal Health, Department of Internal Medicine, Sacramento, California, USA
| | - Yu-An E Lay
- Center for Musculoskeletal Health, Department of Internal Medicine, Sacramento, California, USA
| | - Fernando A Fierro
- Stem Cell Program, UC Davis Health System, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA
| | - Haiyan Chen
- Center for Musculoskeletal Health, Department of Internal Medicine, Sacramento, California, USA.,Adult Programs Division, California Department of Social Services, Sacramento, California, USA
| | - Nancy E Lane
- Center for Musculoskeletal Health, Department of Internal Medicine, Sacramento, California, USA
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Internal Medicine, Sacramento, California, USA
| |
Collapse
|
33
|
Baum R, Gravallese EM. Bone as a Target Organ in Rheumatic Disease: Impact on Osteoclasts and Osteoblasts. Clin Rev Allergy Immunol 2017; 51:1-15. [PMID: 26411424 DOI: 10.1007/s12016-015-8515-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dysregulated bone remodeling occurs when there is an imbalance between bone resorption and bone formation. In rheumatic diseases, including rheumatoid arthritis (RA) and seronegative spondyloarthritis, systemic and local factors disrupt the process of physiologic bone remodeling. Depending upon the local microenvironment, cell types, and local mechanical forces, inflammation results in very different effects on bone, promoting bone loss in the joints and in periarticular and systemic bone in RA and driving bone formation at enthesial and periosteal sites in diseases such as ankylosing spondylitis (AS), included within the classification of axial spondyloarthritis. There has been a great deal of interest in the role of osteoclasts in these processes and much has been learned over the past decade about osteoclast differentiation and function. It is now appreciated that osteoblast-mediated bone formation is also inhibited in the RA joint, limiting the repair of erosions. In contrast, osteoblasts function to produce new bone in AS. The Wnt and BMP signaling pathways have emerged as critical in the regulation of osteoblast function and the outcome for bone in rheumatic diseases, and these pathways have been implicated in both bone loss in RA and bone formation in AS. These pathways provide potential novel approaches for therapeutic intervention in diseases in which inflammation impacts bone.
Collapse
Affiliation(s)
- Rebecca Baum
- Department of Medicine and Division of Rheumatology, University of Massachusetts Medical School, Lazare Research Building Suite 223, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Ellen M Gravallese
- Department of Medicine and Division of Rheumatology, University of Massachusetts Medical School, Lazare Research Building Suite 223, 364 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
34
|
Yao W, Lay YAE, Kot A, Liu R, Zhang H, Chen H, Lam K, Lane NE. Improved Mobilization of Exogenous Mesenchymal Stem Cells to Bone for Fracture Healing and Sex Difference. Stem Cells 2016; 34:2587-2600. [PMID: 27334693 DOI: 10.1002/stem.2433] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/15/2016] [Accepted: 05/06/2016] [Indexed: 01/05/2023]
Abstract
Mesenchymal stem cell (MSC) transplantation has been tested in animal and clinical fracture studies. We have developed a bone-seeking compound, LLP2A-Alendronate (LLP2A-Ale) that augments MSC homing to bone. The purpose of this study was to determine whether treatment with LLP2A-Ale or a combination of LLP2A-Ale and MSCs would accelerate bone healing in a mouse closed fracture model and if the effects are sex dependent. A right mid-femur fracture was induced in two-month-old osterix-mCherry (Osx-mCherry) male and female reporter mice. The mice were subsequently treated with placebo, LLP2A-Ale (500 μg/kg, IV), MSCs derived from wild-type female Osx-mCherry adipose tissue (ADSC, 3 x 105 , IV) or ADSC + LLP2A-Ale. In phosphate buffered saline-treated mice, females had higher systemic and surface-based bone formation than males. However, male mice formed a larger callus and had higher volumetric bone mineral density and bone strength than females. LLP2A-Ale treatment increased exogenous MSC homing to the fracture gaps, enhanced incorporation of these cells into callus formation, and stimulated endochondral bone formation. Additionally, higher engraftment of exogenous MSCs in fracture gaps seemed to contribute to overall fracture healing and improved bone strength. These effects were sex-independent. There was a sex-difference in the rate of fracture healing. ADSC and LLP2A-Ale combination treatment was superior to on callus formation, which was independent of sex. Increased mobilization of exogenous MSCs to fracture sites accelerated endochondral bone formation and enhanced bone tissue regeneration. Stem Cells 2016;34:2587-2600.
Collapse
Affiliation(s)
- Wei Yao
- Department of Internal Medicine, Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, California, USA.
| | - Yu-An Evan Lay
- Department of Internal Medicine, Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, California, USA
| | - Alexander Kot
- Department of Internal Medicine, Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, California, USA
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California at Davis Medical Center, Sacramento, California, USA
| | - Hongliang Zhang
- Department of Internal Medicine, Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, California, USA
| | - Haiyan Chen
- Department of Internal Medicine, Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, California, USA
| | - Kit Lam
- Department of Biochemistry and Molecular Medicine, University of California at Davis Medical Center, Sacramento, California, USA
| | - Nancy E Lane
- Department of Internal Medicine, Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, California, USA
| |
Collapse
|
35
|
The Relationship between Metabolic Syndrome and Osteoporosis: A Review. Nutrients 2016; 8:nu8060347. [PMID: 27338453 PMCID: PMC4924188 DOI: 10.3390/nu8060347] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 12/17/2022] Open
Abstract
Metabolic syndrome (MetS) and osteoporosis are two major healthcare problems worldwide. Metabolic syndrome is a constellation of medical conditions consisting of central obesity, hyperglycemia, hypertension, and dyslipidemia, in which each acts on bone tissue in different ways. The growing prevalence of MetS and osteoporosis in the population along with the controversial findings on the relationship between both conditions suggest the importance for further investigation and discussion on this topic. This review aims to assess the available evidence on the effects of each component of MetS on bone metabolism from the conventional to the contemporary. Previous studies suggested that the two conditions shared some common underlying pathways, which include regulation of calcium homeostasis, receptor activator of NF-κB ligand (RANKL)/receptor activator of the NF-κB (RANK)/osteoprotegerin (OPG) and Wnt-β-catenin signaling pathways. In conclusion, we suggest that MetS may have a potential role in developing osteoporosis and more studies are necessary to further prove this hypothesis.
Collapse
|
36
|
Osteogenesis induced by frizzled-related protein (FRZB) is linked to the netrin-like domain. J Transl Med 2016; 96:570-80. [PMID: 26927515 DOI: 10.1038/labinvest.2016.38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/05/2016] [Accepted: 01/25/2016] [Indexed: 12/17/2022] Open
Abstract
Abnormal Wnt signaling is associated with bone mass disorders. Frizzled-related protein (FRZB, also known as secreted frizzled-related protein-3 (SFRP3)) is a Wnt modulator that contains an amino-terminal cysteine-rich domain (CRD) and a carboxy-terminal Netrin-like (NTN) motif. Frzb(-/-) mice show increased cortical thickness. However, the direct effect of FRZB on osteogenic differentiation and the involvement of the structural domains herein are not fully understood. In this study, we observed that stable overexpression of Frzb in MC3T3-E1 cells increased calcium deposition and osteoblast markers compared with control. Western blot analysis showed that the increased osteogenesis was associated with reduced canonical, but increased non-canonical Wnt signaling. On the contrary, loss of Frzb induced the opposite effects on osteogenesis and Wnt signaling. To translationally validate the positive effects of FRZB on primary human cells, we treated human periosteal and human bone marrow stromal cells with conditioned medium from MC3T3-E1 cells overexpressing Frzb and observed an increase in Alizarin red staining. We further studied the effect of the domains. FrzbNTN overexpression induced similar effects on osteogenesis as full-length Frzb, whereas FrzbCRD overexpressing cells mimicked loss of Frzb experiments. The CRD is considered as the Wnt binding domain, but the NTN domain also has important effects on bone biology. FRZB and other SFRPs or their specific domains may hold surprising potential as therapeutics for bone and joint disorders considering that excess of SFRPs has effects that are not expected under physiological, endogenous expression conditions.
Collapse
|
37
|
Diekwisch TGH. Our periodontal tissue: a masterpiece of evolution. J Clin Periodontol 2016; 43:320-2. [PMID: 26878344 DOI: 10.1111/jcpe.12532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Thomas G H Diekwisch
- Department of Periodontics, Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, TX, USA
| |
Collapse
|
38
|
Li KC, Chang YH, Yeh CL, Hu YC. Healing of osteoporotic bone defects by baculovirus-engineered bone marrow-derived MSCs expressing MicroRNA sponges. Biomaterials 2016; 74:155-66. [DOI: 10.1016/j.biomaterials.2015.09.046] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 12/21/2022]
|
39
|
Shaw AT, Gravallese EM. Mediators of inflammation and bone remodeling in rheumatic disease. Semin Cell Dev Biol 2015; 49:2-10. [PMID: 26481971 DOI: 10.1016/j.semcdb.2015.10.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 10/09/2015] [Indexed: 12/17/2022]
Abstract
Remodeling of bone is a continuous process that occurs throughout life. Under normal physiologic conditions, bone-resorbing osteoclasts and bone-forming osteoblasts are tightly coupled and regulated to ensure proper balance, such that there is no net change in bone mass. However, inflammation perturbs normal bone homeostasis. The impact of inflammation on bone is dependent upon the anatomic site affected, cell types, factors and cytokines present in the local microenvironment, and local mechanical forces. Cytokines are central to the pathogenesis of inflammation-induced bone loss and contribute to the uncoupling of osteoclast-mediated bone resorption and osteoblast-mediated bone formation, thereby disrupting normal remodeling. In this review, we will discuss the effects of cytokines on bone in two settings, rheumatoid arthritis and spondyloarthritis, a disease category that includes ankylosing spondylitis, psoriatic arthritis, reactive arthritis, inflammatory bowel disease, and juvenile onset spondyloarthropathy. The outcome for bone in these disease settings is quite different, and an understanding of the pathogenic mechanisms leading to the net impact on bone has been essential in developing new therapeutic approaches to bone health in these diseases.
Collapse
Affiliation(s)
- Anita T Shaw
- Department of Medicine, Division of Rheumatology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Ellen M Gravallese
- Department of Medicine, Division of Rheumatology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
40
|
Gou Y, Zhang T, Xu J. Transcription Factors in Craniofacial Development: From Receptor Signaling to Transcriptional and Epigenetic Regulation. Curr Top Dev Biol 2015; 115:377-410. [PMID: 26589933 DOI: 10.1016/bs.ctdb.2015.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Craniofacial morphogenesis is driven by spatial-temporal terrains of gene expression, which give rise to stereotypical pattern formation. Transcription factors are key cellular components that control these gene expressions. They are information hubs that integrate inputs from extracellular factors and environmental cues, direct epigenetic modifications, and define transcriptional status. These activities allow transcription factors to confer specificity and potency to transcription regulation during development.
Collapse
Affiliation(s)
- Yongchao Gou
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA
| | - Tingwei Zhang
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA; State Key Laboratory of Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Xu
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA.
| |
Collapse
|
41
|
Visweswaran M, Pohl S, Arfuso F, Newsholme P, Dilley R, Pervaiz S, Dharmarajan A. Multi-lineage differentiation of mesenchymal stem cells - To Wnt, or not Wnt. Int J Biochem Cell Biol 2015; 68:139-47. [PMID: 26410622 DOI: 10.1016/j.biocel.2015.09.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 01/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent precursor cells originating from several adult connective tissues. MSCs possess the ability to self-renew and differentiate into several lineages, and are recognized by the expression of unique cell surface markers. Several lines of evidence suggest that various signal transduction pathways and their interplay regulate MSC differentiation. To that end, a critical player in regulating MSC differentiation is a group of proteins encoded by the Wnt gene family, which was previously known for influencing various stages of embryonic development and cell fate determination. As MSCs have gained significant clinical attention for their potential applications in regenerative medicine, it is imperative to unravel the mechanisms by which molecular regulators control differentiation of MSCs for designing cell-based therapeutics. It is rather coincidental that the functional outcome(s) of Wnt-induced signals share similarities with cellular redox-mediated networks from the standpoint of MSC biology. Furthermore, there is evidence for a crosstalk between Wnt and redox signalling, which begs the question whether Wnt-mediated differentiation signals involve the intermediary role of reactive oxygen species. In this review, we summarize the impact of Wnt signalling on multi-lineage differentiation of MSCs, and attempt to unravel the intricate interplay between Wnt and redox signals.
Collapse
Affiliation(s)
- Malini Visweswaran
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Sebastian Pohl
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Philip Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Rodney Dilley
- Ear Sciences Centre, University of Western Australia and Ear Science Institute Australia, Perth, Western Australia 6008, Australia
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|
42
|
Abstract
BACKGROUND One of the strongest predictors for osteoporosis is peak bone mass. Interventions to augment peak bone mass have yet to be developed. β-Ecdysone (βEcd), a natural steroid-like compound produced by arthropods to initiate metamorphosis, is believed to have androgenic effects and so may be used to augment bone mass. QUESTIONS/PURPOSES The purpose of this study was to use both male and female (1) gonadal-sufficient; and (2) -insufficient mice to investigate sex differences in terms of bone development and structure after βEcd administration. METHODS Two-month-old male and female Swiss-Webster mice were randomized to receive either vehicle or βEcd (0.5 mg/kg) for 3 weeks. In a separate experiment to evaluate the effects of βEcd on sex hormone-deficient mice, gonadectomy was performed in male (orchiectomy [ORX]) and female mice (ovariectomy [OVX]). Sham-operated and the ORX/OVX mice were then treated for 3 weeks with βEcd. Primary endpoints for the study were trabecular bone structure and bone strength. RESULTS In male mice, the trabecular bone volume was 0.18±0.02 in the placebo-treated (PL) and 0.23±0.02 in the βEcd-treated group (p<0.05 versus PL); and 0.09±0.01 in the ORX group (p<0.05 versus PL) and 0.12±0.01 in the ORX+βEcd group. Vertebral bone strength (maximum load) was 43±2 in PL and 51±1 in the βEcd-treated group (p<0.05 versus PL); and 30±4 in the ORX group (p<0.05 versus PL) and 37±3 in the ORX+βEcd group. In female mice, trabecular bone volume was 0.23±0.02 in PL and 0.26±0.02 in the βEcd-treated group (p<0.05 versus PL); and 0.15±0.01 in the OVX group (p<0.05 versus PL) and 0.14±0.01 in the OVX+βEcd group. Maximum load of the vertebrae was 45±2 in PL and 48±4 in the βEcd-treated group; and 39±4 in the OVX group (p<0.05 versus PL) and 44±4 in the OVX+βEcd group. CONCLUSIONS These findings suggest the potential use of βEcd in the augmentation of bone mass in growing male and female mice. It may also partially prevent the detrimental effects of gonadectomy on trabecular bone. CLINICAL RELEVANCE Our results support the potential use of βEcd or nature products that are rich in βEcd to augment peak bone mass. βEcd may differ from the other anabolic hormone treatments that may have severe side effects such as serious cardiac complications. However, its effects on humans remain to be determined.
Collapse
|
43
|
Dai W, Jiang L, Lay YAE, Chen H, Jin G, Zhang H, Kot A, Ritchie RO, Lane NE, Yao W. Prevention of glucocorticoid induced bone changes with beta-ecdysone. Bone 2015; 74:48-57. [PMID: 25585248 PMCID: PMC4355031 DOI: 10.1016/j.bone.2015.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/15/2014] [Accepted: 01/05/2015] [Indexed: 12/20/2022]
Abstract
Beta-ecdysone (βEcd) is a phytoecdysteroid found in the dry roots and seeds of the asteraceae and achyranthes plants, and is reported to increase osteogenesis in vitro. Since glucocorticoid (GC) excess is associated with a decrease in bone formation, the purpose of this study was to determine if treatment with βEcd could prevent GC-induced osteoporosis. Two-month-old male Swiss-Webster mice (n=8-10/group) were randomized to either placebo or slow release prednisolone pellets (3.3mg/kg/day) and treated with vehicle control or βEcd (0.5mg/kg/day) for 21days. GC treatment inhibited age-dependent trabecular gain and cortical bone expansion and this was accompanied by a 30-50% lower bone formation rate (BFR) at both the endosteal and periosteal surfaces. Mice treated with only βEcd significantly increased bone formation on the endosteal and periosteal bone surfaces, and increased cortical bone mass were their controls to compare to GC alone. Concurrent treatment of βEcd and GC completely prevented the GC-induced reduction in BFR, trabecular bone volume and partially prevented cortical bone loss. In vitro studies determined that βEcd prevented the GC increase in autophagy of the bone marrow stromal cells as well as in whole bone. In summary, βEcd prevented GC induced changes in bone formation, bone cell viability and bone mass. Additional studies are warranted of βEcd for the treatment of GC induced bone loss.
Collapse
Affiliation(s)
- Weiwei Dai
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA; Department of Science and Technology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Li Jiang
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Yu-An Evan Lay
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Haiyan Chen
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Guoqin Jin
- Department of Science and Technology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Hongliang Zhang
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Alexander Kot
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Robert O Ritchie
- Department of Materials Science and Engineering, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Nancy E Lane
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Wei Yao
- Center for Musculoskeletal Health, Internal Medicine, University of California at Davis Medical Center, Sacramento, CA 95817, USA.
| |
Collapse
|
44
|
Tian L, Yu X. Lipid metabolism disorders and bone dysfunction--interrelated and mutually regulated (review). Mol Med Rep 2015; 12:783-94. [PMID: 25760577 PMCID: PMC4438959 DOI: 10.3892/mmr.2015.3472] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 12/03/2014] [Indexed: 02/05/2023] Open
Abstract
The association between lipid and bone metabolism has become an increasing focus of interest in recent years, and accumulating evidence has shown that atherosclerosis (AS) and osteoporosis (OP), a disorder of bone metabolism, frequently co-exist. Fat and bone are known to share a common progenitor cell: Multipotent mesenchymal stem cells (MSC) in the bone marrow (BM), which are able to differentiate into various cell phenotypes, including osteoblasts, adipocytes and chondrocytes. Laboratory-based and clinical trials have shown that increasing adipocytes are accompanied by a decrease in bone mineral density (BMD) and bone mass. Statins, lipid-lowering drugs used to treat hyperlipidemia, also provide benefit in the treatment of OP. There is thus evidence that the metabolism of lipids is correlated with that of bone, and that the two are mutually regulated. The present review primarily focuses on the potential association between lipid metabolism disturbance and OP, based on biological metabolism, pathophysiological processes, results from clinical and experimental animal studies, processes involved in the differentiation of adipocytes and osteoblasts, as well as pharmacological treatments of these diseases.
Collapse
Affiliation(s)
- Li Tian
- Laboratory of Endocrinology and Metabolism, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
45
|
Zhang L, Chen P, Chen L, Weng T, Zhang S, Zhou X, Zhang B, Liu L. Inhibited Wnt signaling causes age-dependent abnormalities in the bone matrix mineralization in the Apert syndrome FGFR2(S252W/+) mice. PLoS One 2015; 10:e112716. [PMID: 25693202 PMCID: PMC4333342 DOI: 10.1371/journal.pone.0112716] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 10/14/2014] [Indexed: 01/16/2023] Open
Abstract
Apert syndrome (AS) is a type of autosomal dominant disease characterized by premature fusion of the cranial sutures, severe syndactyly, and other abnormalities in internal organs. Approximately 70% of AS cases are caused by a single mutation, S252W, in fibroblast growth factor receptor 2 (FGFR2). Two groups have generated FGFR2 knock-in mice Fgfr2S252W/+ that exhibit features of AS. During the present study of AS using the Fgfr2S252W/+ mouse model, an age-related phenotype of bone homeostasis was discovered. The long bone mass was lower in 2 month old mutant mice than in age-matched controls but higher in 5 month old mutant mice. This unusual phenotype suggested that bone marrow-derived mesenchymal stem cells (BMSCs), which are vital to maintain bone homeostasis, might be involved. BMSCs were isolated from Fgfr2S252W/+ mice and found that S252W mutation could impair osteogenic differentiation BMSCs but enhance mineralization of more mature osteoblasts. A microarray analysis revealed that Wnt pathway inhibitors SRFP1/2/4 were up-regulated in mutant BMSCs. This work provides evidence to show that the Wnt/β-catenin pathway is inhibited in both mutant BMSCs and osteoblasts, and differentiation defects of these cells can be ameliorated by Wnt3a treatment. The present study suggested that the bone abnormalities caused by deregulation of Wnt pathway may underlie the symptoms of AS.
Collapse
Affiliation(s)
- Li Zhang
- Department of Stomatology, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
- Department 4, Daping Hospital & Research Institute of Surgery, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | - Peng Chen
- Department 4, Daping Hospital & Research Institute of Surgery, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing 400042, China
- Neurosurgery Department, PLA 324 Hospital, Chongqing, China
| | - Lin Chen
- Department of Stomatology, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Tujun Weng
- Department 4, Daping Hospital & Research Institute of Surgery, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | - Shichang Zhang
- Department 4, Daping Hospital & Research Institute of Surgery, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | - Xia Zhou
- Department of Stomatology, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Bo Zhang
- Department 4, Daping Hospital & Research Institute of Surgery, Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | - Luchuan Liu
- Department of Stomatology, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
46
|
Vanderschueren D, Laurent MR, Claessens F, Gielen E, Lagerquist MK, Vandenput L, Börjesson AE, Ohlsson C. Sex steroid actions in male bone. Endocr Rev 2014; 35:906-60. [PMID: 25202834 PMCID: PMC4234776 DOI: 10.1210/er.2014-1024] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sex steroids are chief regulators of gender differences in the skeleton, and male gender is one of the strongest protective factors against osteoporotic fractures. This advantage in bone strength relies mainly on greater cortical bone expansion during pubertal peak bone mass acquisition and superior skeletal maintenance during aging. During both these phases, estrogens acting via estrogen receptor-α in osteoblast lineage cells are crucial for male cortical and trabecular bone, as evident from conditional genetic mouse models, epidemiological studies, rare genetic conditions, genome-wide meta-analyses, and recent interventional trials. Genetic mouse models have also demonstrated a direct role for androgens independent of aromatization on trabecular bone via the androgen receptor in osteoblasts and osteocytes, although the target cell for their key effects on periosteal bone formation remains elusive. Low serum estradiol predicts incident fractures, but the highest risk occurs in men with additionally low T and high SHBG. Still, the possible clinical utility of serum sex steroids for fracture prediction is unknown. It is likely that sex steroid actions on male bone metabolism rely also on extraskeletal mechanisms and cross talk with other signaling pathways. We propose that estrogens influence fracture risk in aging men via direct effects on bone, whereas androgens exert an additional antifracture effect mainly via extraskeletal parameters such as muscle mass and propensity to fall. Given the demographic trends of increased longevity and consequent rise of osteoporosis, an increased understanding of how sex steroids influence male bone health remains a high research priority.
Collapse
Affiliation(s)
- Dirk Vanderschueren
- Clinical and Experimental Endocrinology (D.V.) and Gerontology and Geriatrics (M.R.L., E.G.), Department of Clinical and Experimental Medicine; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine (M.R.L., F.C.); and Centre for Metabolic Bone Diseases (D.V., M.R.L., E.G.), KU Leuven, B-3000 Leuven, Belgium; and Center for Bone and Arthritis Research (M.K.L., L.V., A.E.B., C.O.), Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR, Vogel P. High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res 2014; 2:14034. [PMID: 26273529 PMCID: PMC4472125 DOI: 10.1038/boneres.2014.34] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Screening gene function in vivo is a powerful approach to discover novel drug targets. We present high-throughput screening (HTS) data for 3 762 distinct global gene knockout (KO) mouse lines with viable adult homozygous mice generated using either gene-trap or homologous recombination technologies. Bone mass was determined from DEXA scans of male and female mice at 14 weeks of age and by microCT analyses of bones from male mice at 16 weeks of age. Wild-type (WT) cagemates/littermates were examined for each gene KO. Lethality was observed in an additional 850 KO lines. Since primary HTS are susceptible to false positive findings, additional cohorts of mice from KO lines with intriguing HTS bone data were examined. Aging, ovariectomy, histomorphometry and bone strength studies were performed and possible non-skeletal phenotypes were explored. Together, these screens identified multiple genes affecting bone mass: 23 previously reported genes (Calcr, Cebpb, Crtap, Dcstamp, Dkk1, Duoxa2, Enpp1, Fgf23, Kiss1/Kiss1r, Kl (Klotho), Lrp5, Mstn, Neo1, Npr2, Ostm1, Postn, Sfrp4, Slc30a5, Slc39a13, Sost, Sumf1, Src, Wnt10b), five novel genes extensively characterized (Cldn18, Fam20c, Lrrk1, Sgpl1, Wnt16), five novel genes with preliminary characterization (Agpat2, Rassf5, Slc10a7, Slc26a7, Slc30a10) and three novel undisclosed genes coding for potential osteoporosis drug targets.
Collapse
Affiliation(s)
| | - Jeff Liu
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| | | | | | | | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals , The Woodlands, TX, USA
| |
Collapse
|
48
|
Saidak Z, Le Henaff C, Azzi S, Marty C, Marie PJ. Low-dose PTH increases osteoblast activity via decreased Mef2c/Sost in senescent osteopenic mice. J Endocrinol 2014; 223:25-33. [PMID: 25056116 DOI: 10.1530/joe-14-0249] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intermittent administration of parathyroid hormone (PTH) 1-34 at a standard dose has been shown to induce anabolic effects in bone. However, whether low-dose PTH promotes bone formation during senescence is unknown. To address this issue, we determined the effects of low-dose PTH and analysed the underlying mechanisms in prematurely senescent mice that display osteopenia. Treatment of 9-week-old Samp6 mice for 6 weeks with PTH at a standard dose (100 μg/kg per day) increased vertebral and femoral bone mass and improved bone microarchitecture as a result of increased bone-forming surfaces and mineral apposition rate (MAR). At a tenfold lower dose (10 μg/kg per day), PTH increased axial bone volume and trabecular thickness, as detected by bone histomorphometry but not by micro-computed tomography analysis. This anabolic effect resulted from increased osteoblast activity, as reflected by increased serum N-terminal propeptide of type 1 procollagen (P1NP) levels and MAR, with unchanged bone-forming surface or osteoblast surface. Mechanistically, low-dose PTH increased the expression of osteoblast markers in bone marrow stromal cells and mature osteoblasts, which was associated with increased expression of the Wnt effector Wisp1. Moreover, low-dose PTH decreased the expression of the Mef2c transcription factor, resulting in decreased Sost expression in osteoblasts/osteocytes. These results indicate that PTH at a low dose is effective at promoting bone formation and increased bone volume in senescent osteopenic mice through increased osteoblast activity and modulation of specific Wnt effectors, which raises the potential therapeutic use of intermittent PTH at low dose to increase bone forming activity and bone mass in skeletal senescence.
Collapse
Affiliation(s)
- Zuzana Saidak
- UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France
| | - Carole Le Henaff
- UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France
| | - Sofia Azzi
- UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France
| | - Caroline Marty
- UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France
| | - Pierre J Marie
- UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France UMR-1132 InsermHôpital Lariboisière, 2 Rue Ambroise Paré, 75475 Paris Cedex 10, FranceUniversité Paris DiderotSorbonne Paris Cité, Paris, France
| |
Collapse
|
49
|
Lee TC, Burghardt AJ, Yao W, Lane NE, Majumdar S, Gullberg GT, Seo Y. Improved trabecular bone structure of 20-month-old male spontaneously hypertensive rats. Calcif Tissue Int 2014; 95:282-91. [PMID: 25106873 PMCID: PMC4153466 DOI: 10.1007/s00223-014-9893-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 07/03/2014] [Indexed: 02/07/2023]
Abstract
A few clinical studies have reported that elderly male participants with hypertensive disease frequently have higher bone mineral density (BMD) than the normotensive participants at several skeletal sites. The detailed mechanism is still unknown; therefore, a study of bone structure and density using the hypertensive animal models could be informative. We used micro-computed tomography to quantitatively evaluate the tibial and 3rd lumbar vertebral bones in the 20-month-old male spontaneous hypertensive rat (SHR). The BMD, volume fraction, and the microarchitecture changes of the SHR were compared to those of same-age normotensive controls (Wistar-Kyoto rat, WKY). We found that in the very old (20 month) male rats, the trabecular bone fraction and microstructure were higher than those in the same-age normotensive controls. The observation of the association of hypertension with BMD and bone strength in hypertensive rats warrants further investigations of bone mass and strength in elderly males with hypertension.
Collapse
Affiliation(s)
- Tzu-Cheng Lee
- Physics Research Laboratory, Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, United States of America
| | - Andrew J. Burghardt
- Musculoskeletal Quantitative Imaging Research Group, Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, United States of America
| | - Wei Yao
- Center for Musculoskeletal Health, Department of Medicine, University of California at Davis, Sacramento, California, United States of America
| | - Nancy E. Lane
- Center for Musculoskeletal Health, Department of Medicine, University of California at Davis, Sacramento, California, United States of America
| | - Sharmila Majumdar
- Musculoskeletal Quantitative Imaging Research Group, Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, United States of America
| | - Grant T. Gullberg
- Department of Radiotracer Development & Imaging Technology, Life Science Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Youngho Seo
- Physics Research Laboratory, Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, United States of America
- Corresponding author: Physics Research Laboratory, China Basin Landing, Lobby 6, Suite 350, 185 Berry St, University of California, San Francisco, CA 94143, USA. Phone: +1 415-353-9464,
| |
Collapse
|
50
|
Yao W, Guan M, Jia J, Dai W, Lay YAE, Amugongo S, Liu R, Olivos D, Saunders M, Lam KS, Nolta J, Olvera D, Ritchie RO, Lane NE. Reversing bone loss by directing mesenchymal stem cells to bone. Stem Cells 2014; 31:2003-14. [PMID: 23818248 DOI: 10.1002/stem.1461] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/01/2013] [Accepted: 03/26/2013] [Indexed: 01/16/2023]
Abstract
Bone regeneration by systemic transplantation of mesenchymal stem cells (MSCs) is problematic due to the inability to control the MSCs' commitment, growth, and differentiation into functional osteoblasts on the bone surface. Our research group has developed a method to direct the MSCs to the bone surface by conjugating a synthetic peptidomimetic ligand (LLP2A) that has high affinity for activated α4β1 integrin on the MSC surface, with a bisphosphonates (alendronate) that has high affinity for bone (LLP2A-Ale), to direct the transplanted MSCs to bone. Our in vitro experiments demonstrated that mobilization of LLP2A-Ale to hydroxyapatite accelerated MSC migration that was associated with an increase in the phosphorylation of Akt kinase and osteoblastogenesis. LLP2A-Ale increased the homing of the transplanted MSCs to bone as well as the osteoblast surface, significantly increased the rate of bone formation and restored both trabecular and cortical bone loss induced by estrogen deficiency or advanced age in mice. These results support LLP2A-Ale as a novel therapeutic option to direct the transplanted MSCs to bone for the treatment of established bone loss related to hormone deficiency and aging.
Collapse
Affiliation(s)
- Wei Yao
- Department of Internal Medicine, University of California at Davis Medical Center, Sacramento, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|