1
|
Nguyen TK, Pham DV, Park PH. Leptin impairs the therapeutic efficacy of adipose-derived mesenchymal stem cells by inducing apoptosis through NLRP3 inflammasomes activation. Biochem Pharmacol 2025; 236:116868. [PMID: 40081766 DOI: 10.1016/j.bcp.2025.116868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/11/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Mesenchymal stem cells (MSC) have been widely applied for regenerative medicine and the treatment of immune-disorders due to their multilineage differentiation and potent immunomodulatory properties. The therapeutic application of MSC post transplantation are influenced by various endogenous modulators. Leptin, a hormone primarily derived from adipose tissue, exerts a variety of physiological functions, in addition to the metabolic effects. In this study, we examined the effects of leptin on the viability of adipose-derived mesenchymal stem cells (ADSC) and its underlying molecular mechanisms with a particular focus on NLRP3 inflammasomes, which serve as signaling platform of the innate immune system. Leptin significantly decreased the viability of ADSC and induced apoptosis. Mechanistically, NLRP3 inflammasomes signaling critically contributes to leptin-induced apoptosis of ADSC by upregulating p53 and Puma. In addition, NLRP3 inflammasomes activation by leptin is mediated via ER stress induction and ROS accumulation. Finally, suppression of ADSC therapeutic efficacy by leptin and the critical role of NLRP3 inflammasomes in this phenomenon were confirmed in DSS-induced colitis model. Pre-conditioning with leptin before transplantation impaired the therapeutic efficacy and immunomodulatory function of ADSC, which were restored by treatment with a pharmacological inhibitor of NLRP3 inflammasomes. Taken together, the results suggest that leptin induces apoptotic cell death in ADSC and impairs the therapeutic effectiveness of ADSC by activating NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Thi-Kem Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Duc-Vinh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Department of Pharmacology, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, South Korea.
| |
Collapse
|
2
|
Li Y, Li P, Xue K, Shi P, Xie X, Wang J, Xu C. LepR-Expressing Cells in Bone and Periodontium. Oral Dis 2025; 31:1065-1072. [PMID: 39748446 DOI: 10.1111/odi.15211] [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: 06/23/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE LepR-expressing cells (LepR+ cells), a critical subpopulation of mesenchymal stem cells, have gained increasing attention in the last decade. LepR+ cells have been found to play a crucial role in maintaining bone and periodontal homeostasis. This review summarizes current research advances focusing on the role of LepR+ cells and their underlying regulatory molecular mechanisms in bones and periodontium, aiming to provide a better understanding of the therapeutic potential of this cell lineage. METHODS A literature review was conducted based on publications in PubMed over the past 20 years, summarizing the research progress on LepR+ cells in bone and periodontal tissues. RESULTS Current evidence revealed that LepR+ cells possess the ability of self-renewal and multilineage differentiation and are essential for bone turnover and periodontal tissue remodeling. In addition, LepR+ cells participate in the processes of bone fracture healing and alveolar socket healing. Moreover, under pathological conditions such as osteoporosis, bone marrow fibrosis, and periodontitis, LepR+ cells exhibit enhanced adipogenic or fibrogenic differentiation abilities. CONCLUSION Therapeutic approaches targeting the cell fate of LepR+ cells hold the potential to provide novel insights into bone/periodontal repair and regeneration therapy.
Collapse
Affiliation(s)
- Yue Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Peitong Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kun Xue
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Peilei Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xudong Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunmei Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Xiang Q, Wu Z, Zhao Y, Tian S, Lin J, Wang L, Jiang S, Sun Z, Li W. Cellular and molecular mechanisms underlying obesity in degenerative spine and joint diseases. Bone Res 2024; 12:71. [PMID: 39658574 PMCID: PMC11632072 DOI: 10.1038/s41413-024-00388-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/22/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
Degenerative spine and joint diseases, including intervertebral disc degeneration (IDD), ossification of the spinal ligaments (OSL), and osteoarthritis (OA), are common musculoskeletal diseases that cause pain or disability to the patients. However, the pathogenesis of these musculoskeletal disorders is complex and has not been elucidated clearly to date. As a matter of fact, the spine and joints are not independent of other organs and tissues. Recently, accumulating evidence demonstrates the association between obesity and degenerative musculoskeletal diseases. Obesity is a common metabolic disease characterized by excessive adipose tissue or abnormal adipose distribution in the body. Excessive mechanical stress is regarded as a critical risk factor for obesity-related pathology. Additionally, obesity-related factors, mainly including lipid metabolism disorder, dysregulated pro-inflammatory adipokines and cytokines, are reported as plausible links between obesity and various human diseases. Importantly, these obesity-related factors are deeply involved in the regulation of cell phenotypes and cell fates, extracellular matrix (ECM) metabolism, and inflammation in the pathophysiological processes of degenerative spine and joint diseases. In this study, we systematically discuss the potential cellular and molecular mechanisms underlying obesity in these degenerative musculoskeletal diseases, and hope to provide novel insights for developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Qian Xiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Zhenquan Wu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Yongzhao Zhao
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Shuo Tian
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Jialiang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Longjie Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Shuai Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Zhuoran Sun
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
| |
Collapse
|
4
|
Graef F, Wei Y, Garbe A, Seemann R, Zenzes M, Tsitsilonis S, Duda GN, Zaslansky P. Increased cancellous bone mass accompanies decreased cortical bone mineral density and higher axial deformation in femurs of leptin-deficient obese mice. J Mech Behav Biomed Mater 2024; 160:106745. [PMID: 39317095 DOI: 10.1016/j.jmbbm.2024.106745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Leptin is a pleiotropic hormone that regulates food intake and energy homeostasis with enigmatic effects on bone development. It is unclear if leptin promotes or inhibits bone growth. The aim of this study was to characterize the micro-architecture and mechanical competence of femur bones of leptin-deficient mice. MATERIALS AND METHODS Right femur bones of 15-week old C57BL/6 (n = 9) and leptin-deficient (ob/ob, n = 9) mice were analyzed. Whole bones were scanned using micro-CT and morphometric parameters of the cortex and trabeculae were assessed. Elastic moduli were determined from microindentations in midshaft cross-sections. Mineral densities were determined using quantitative backscatter scanning electron microscopy. 3D models of the distal femur metaphysis, cleared from trabecular bone, were meshed and used for finite element simulations of axial loading to identify straining differences between ob/ob and C57BL/6 controls. RESULTS Compared with C57BL/6 controls, ob/ob mice had significantly shorter bones. ob/ob mice showed significantly increased cancellous bone volume and trabecular thickness. qBEI quantified a ∼7% lower mineral density in ob/ob mice in the distal femur metaphysis. Indentation demonstrated a significantly reduced Young's modulus of 12.14 [9.67, 16.56 IQR] GPa for ob/ob mice compared to 23.12 [20.70, 26.57 IQR] GPa in C57BL/6 mice. FEA revealed greater deformation of cortical bone in ob/ob as compared to C57BL/6 mice. CONCLUSION Leptin deficient ob/ob mice have a softer cortical bone in the distal femur metaphysis but an excessive amount of cancellous bone, possibly as a response to increased deformation of the bones during axial loading. Both FEA and direct X-ray and electron microscopy imaging suggest that the morphology and micro-architecture of ob/ob mice have inferior biomechanical properties suggestive of a reduced mechanical competence.
Collapse
Affiliation(s)
- F Graef
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany; Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany.
| | - Y Wei
- Charité - Universitätsmedizin Berlin, Department of Operative and Preventive Dentistry, Germany; Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany.
| | - A Garbe
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany
| | - R Seemann
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany
| | - M Zenzes
- Charité - Universitätsmedizin Berlin, Department of Operative and Preventive Dentistry, Germany
| | - S Tsitsilonis
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany; Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - G N Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - P Zaslansky
- Charité - Universitätsmedizin Berlin, Department of Operative and Preventive Dentistry, Germany.
| |
Collapse
|
5
|
Liu Z, Xie W, Li H, Liu X, Lu Y, Lu B, Deng Z, Li Y. Novel perspectives on leptin in osteoarthritis: Focus on aging. Genes Dis 2024; 11:101159. [PMID: 39229323 PMCID: PMC11369483 DOI: 10.1016/j.gendis.2023.101159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/10/2023] [Accepted: 09/16/2023] [Indexed: 09/05/2024] Open
Abstract
Osteoarthritis (OA) is a common chronic joint disease characterized by articular cartilage degeneration, subchondral sclerosis, synovitis, and osteophyte formation. OA is associated with disability and impaired quality of life, particularly among the elderly. Leptin, a 16-kD non-glycosylated protein encoded by the obese gene, is produced on a systemic and local basis in adipose tissue and the infrapatellar fat pad located in the knee. The metabolic mechanisms employed by leptin in OA development have been widely studied, with attention being paid to aging as a corroborative risk factor for OA. Hence, in this review, we have attempted to establish a potential link between leptin and OA, by focusing on aging-associated mechanisms and proposing leptin as a potential diagnostic and therapeutic target in aging-related mechanisms of OA that may provide fruitful guidance and emphasis for future research.
Collapse
Affiliation(s)
- Zimo Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xu Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhenhan Deng
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
6
|
Li J, Zhang Z, Tang J, Hou Z, Li L, Li B. Emerging roles of nerve-bone axis in modulating skeletal system. Med Res Rev 2024; 44:1867-1903. [PMID: 38421080 DOI: 10.1002/med.22031] [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: 12/04/2023] [Revised: 01/25/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Over the past decades, emerging evidence in the literature has demonstrated that the innervation of bone is a crucial modulator for skeletal physiology and pathophysiology. The nerve-bone axis sparked extensive preclinical and clinical investigations aimed at elucidating the contribution of nerve-bone crosstalks to skeleton metabolism, homeostasis, and injury repair through the perspective of skeletal neurobiology. To date, peripheral nerves have been widely reported to mediate bone growth and development and fracture healing via the secretion of neurotransmitters, neuropeptides, axon guidance factors, and neurotrophins. Relevant studies have further identified several critical neural pathways that stimulate profound alterations in bone cell biology, revealing a complex interplay between the skeleton and nerve systems. In addition, inspired by nerve-bone crosstalk, novel drug delivery systems and bioactive materials have been developed to emulate and facilitate the process of natural bone repair through neuromodulation, eventually boosting osteogenesis for ideal skeletal tissue regeneration. Overall, this work aims to review the novel research findings that contribute to deepening the current understanding of the nerve-bone axis, bringing forth some schemas that can be translated into the clinical scenario to highlight the critical roles of neuromodulation in the skeletal system.
Collapse
Affiliation(s)
- Jingya Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhuoyuan Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinru Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zeyu Hou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Pham DV, Nguyen TK, Park PH. Adipokines at the crossroads of obesity and mesenchymal stem cell therapy. Exp Mol Med 2023; 55:313-324. [PMID: 36750692 PMCID: PMC9981593 DOI: 10.1038/s12276-023-00940-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 02/09/2023] Open
Abstract
Mesenchymal stem cell (MSC) therapy is an emerging treatment strategy to counteract metabolic syndromes, including obesity and its comorbid disorders. However, its effectiveness is challenged by various factors in the obese environment that negatively impact MSC survival and function. The identification of these detrimental factors will provide opportunities to optimize MSC therapy for the treatment of obesity and its comorbidities. Dysregulated production of adipokines, a group of cytokines and hormones derived from adipose tissue, has been postulated to play a pivotal role in the development of obesity-associated complications. Intriguingly, adipokines have also been implicated in the modulation of viability, self-renewal, proliferation, and other properties of MSC. However, the involvement of adipokine imbalance in impaired MSC functionality has not been completely understood. On the other hand, treatment of obese individuals with MSC can restore the serum adipokine profile, suggesting the bidirectionality of the adipokine-MSC relationship. In this review, we aim to discuss the current knowledge on the central role of adipokines in the crosstalk between obesity and MSC dysfunction. We also summarize recent advances in the use of MSC for the treatment of obesity-associated diseases to support the hypothesis that adipokines modulate the benefits of MSC therapy in obese patients.
Collapse
Affiliation(s)
- Duc-Vinh Pham
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Thi-Kem Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea. .,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
8
|
Zhuang Y, Yang W, Zhang L, Fan C, Qiu L, Zhao Y, Chen B, Chen Y, Shen H, Dai J. A novel leptin receptor binding peptide tethered-collagen scaffold promotes lung injury repair. Biomaterials 2022; 291:121884. [DOI: 10.1016/j.biomaterials.2022.121884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/10/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
|
9
|
Zhang Z, Zhou H, Sun F, Han J, Han Y. Circ_FBLN1 promotes the proliferation and osteogenic differentiation of human bone marrow-derived mesenchymal stem cells by regulating let-7i-5p/FZD4 axis and Wnt/β-catenin pathway. J Bioenerg Biomembr 2021; 53:561-572. [PMID: 34424449 DOI: 10.1007/s10863-021-09917-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/13/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Recently, more and more circular RNAs (circRNAs) have been identified in osteogenesis. In this study, we aimed to explore the effect of circ_FBLN1 on the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs). METHODS The protein levels of osteogenesis-related genes, let-7i-5p, frizzled class receptor 4 (FZD4), Ki67, Wnt6 and β-catenin were measured by western blot assay. The levels of circ_FBLN1, FBLN1 mRNA and FZD4 mRNA were determined by quantitative real-time polymerase chain reaction (qRT-PCR) assay. The feature of circ_FBLN1 was investigated by RNase R and Actinomycin D assays. Cell proliferation ability was evaluated by colony formation assay and 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay. The targeting relationship between let-7i-5p and circ_FBLN1 or FZD4 was verified by dual-luciferase reporter assay. RESULTS Circ_FBLN1 level was enhanced during the osteogenic differentiation of hBMSCs. Silencing of circ_FBLN1 repressed cell proliferation and osteogenic differentiation in hBMSCs. For mechanism analysis, circ_FBLN1 was found to act as a sponge for let-7i-5p and FZD4 served as a direct target gene of let-7i-5p. Let-7i-5p was downregulated during the osteogenic differentiation of hBMSCs and let-7i-5p inhibition restored the effects of circ_FBLN1 knockdown on the proliferation and osteogenesis of hBMSCs. Moreover, let-7i-5p overexpression suppressed cell proliferation and osteogenesis in hBMSCs through targeting FZD4. In addition, circ_FBLN1 knockdown reduced the levels of Wnt6 and β-catenin in hBMSCs, indicating the inactivation of Wnt/β-catenin pathway. CONCLUSION Knockdown of circ_FBLN1 inhibited the proliferation and osteogenesis of hBMSCs by regulating let-7i-5p/FZD4 axis and repressing Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Zilong Zhang
- Department of Spine, Zaozhuang Municipal Hospital, Zaozhuang City, Shandong Province, China
| | - Huachao Zhou
- Department of the Orthopaedic Trauma, Zaozhuang Mining Group Zaozhuang Hospital, Zaozhuang, Shandong, China
| | - Fei Sun
- Department of the Orthopaedic Trauma, Zaozhuang Mining Group Zaozhuang Hospital, Zaozhuang, Shandong, China
| | - Jianjian Han
- Department of the Orthopaedic Trauma, Qingdao Central Hospital, Qingdao, Shandong, China
| | - Yongyuan Han
- Department of Orthopedics, No.4 Hospital Beijing University of Chinese Medicine, No.202 Xuezhuang Community, Zhongxin Street, Zaozhuang, 277101, Shandong, China.
| |
Collapse
|
10
|
Kowal JM, Möller S, Ali D, Figeac F, Barington T, Schmal H, Kassem M. Identification of a clinical signature predictive of differentiation fate of human bone marrow stromal cells. Stem Cell Res Ther 2021; 12:265. [PMID: 33941262 PMCID: PMC8091554 DOI: 10.1186/s13287-021-02338-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/19/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Transplantation of human bone marrow stromal cells (hBMSCs) is a promising therapy for bone regeneration due to their ability to differentiate into bone forming osteoblastic cells. However, transplanted hBMSCs exhibit variable capacity for bone formation resulting in inconsistent clinical outcome. The aim of the study was to identify a set of donor- and cell-related characteristics that detect hBMSCs with optimal osteoblastic differentiation capacity. METHODS We collected hBMSCs from 58 patients undergoing surgery for bone fracture. Clinical profile of the donors and in vitro characteristics of cultured hBMSCs were included in uni- and multivariable analysis to determine their predictive value for osteoblastic versus adipocytic differentiation capacity assessed by quantification of mineralized matrix and mature adipocyte formation, respectively. RESULTS We identified a signature that explained > 50% of variation in osteoblastic differentiation outcome which included the following positive predictors: donor sex (male), absence of osteoporosis diagnosis, intake of vitamin D supplements, higher fraction of CD146+, and alkaline phosphate (ALP+) cells. With the exception of vitamin D and ALP+ cells, these variables were also negative predictors of adipocytic differentiation. CONCLUSIONS Using a combination of clinical and cellular criteria, it is possible to predict differentiation outcome of hBMSCs. This signature may be helpful in selecting donor cells in clinical trials of bone regeneration.
Collapse
Affiliation(s)
- Justyna Magdalena Kowal
- Department of Endocrinology, Odense University Hospital, Odense, Denmark. .,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Sören Möller
- OPEN - Open Patient data Explorative Network, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Dalia Ali
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Florence Figeac
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Torben Barington
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Hagen Schmal
- Department of Orthopedics and Traumatology, Odense University Hospital, Odense, Denmark.,Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Moustapha Kassem
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.,Molecular Endocrinology Unit (KMEB), Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200, Copenhagen, Denmark
| |
Collapse
|
11
|
Martyniak K, Wei F, Ballesteros A, Meckmongkol T, Calder A, Gilbertson T, Orlovskaya N, Coathup MJ. Do polyunsaturated fatty acids protect against bone loss in our aging and osteoporotic population? Bone 2021; 143:115736. [PMID: 33171312 DOI: 10.1016/j.bone.2020.115736] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023]
Abstract
Age-related bone loss is inevitable in both men and women and there will soon be more people of extreme old age than ever before. Osteoporosis is a common chronic disease and as the proportion of older people, rate of obesity and the length of life increases, a rise in age-related degenerating bone diseases, disability, and prolonged dependency is projected. Fragility fractures are one of the most severe complications associated with both primary and secondary osteoporosis and current treatment strategies target weight-bearing exercise and pharmacological intervention, both with limited long-term success. Obesity and osteoporosis are intimately interrelated, and diet is a variable that plays a significant role in bone regeneration and repair. The Western Diet is characterized by its unhealthy components, specifically excess amounts of saturated fat intake. This review examines the impact of saturated and polyunsaturated fatty acid consumption on chronic inflammation, osteogenesis, bone architecture, and strength and explores the hypothesis that dietary polyunsaturated fats have a beneficial effect on osteogenesis, reducing bone loss by decreasing chronic inflammation, and activating bone resorption through key cellular and molecular mechanisms in our aging population. We conclude that aging, obesity and a diet high in saturated fatty acids significantly impairs bone regeneration and repair and that consumption of ω-3 polyunsaturated fatty acids is associated with significantly increased bone regeneration, improved microarchitecture and structural strength. However, ω-6 polyunsaturated fatty acids were typically pro-inflammatory and have been associated with an increased fracture risk. This review suggests a potential role for ω-3 fatty acids as a non-pharmacological dietary method of reducing bone loss in our aging population. We also conclude that contemporary amendments to the formal nutritional recommendations made by the Food and Nutrition Board may be necessary such that our aging population is directly considered.
Collapse
Affiliation(s)
- Kari Martyniak
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Amelia Ballesteros
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Teerin Meckmongkol
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Department of General Surgery, Nemours Children's Hospital, Orlando, FL, United States
| | - Ashley Calder
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Timothy Gilbertson
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Nina Orlovskaya
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, United States
| | - Melanie J Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States.
| |
Collapse
|
12
|
Feng H, Zhang Q, Zhao Y, Zhao L, Shan B. Leptin acts on mesenchymal stem cells to promote chemoresistance in osteosarcoma cells. Aging (Albany NY) 2020; 12:6340-6351. [PMID: 32289750 PMCID: PMC7185129 DOI: 10.18632/aging.103027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 02/27/2020] [Indexed: 04/17/2023]
Abstract
Leptin signaling influences osteoblastogenesis and modulates the fate of mesenchymal stem cells (MSCs) during bone and cartilage regeneration. Although MSCs abound in the osteosarcoma (OS) microenvironment, and leptin exhibits pro-tumorigenic properties, leptin's influence on OS progression and chemoresistant signaling in MSCs remains unclear. Using cell viability and apoptosis assays, we showed that medium conditioned by leptin-treated human MSCs promotes cisplatin resistance in cultured human OS cells. Moreover, GFP-LC3 expression and chloroquine treatment experiments showed that this effect is mediated by stimulation of autophagy in OS cells. TGF-β expression in MSCs was upregulated by leptin and suppressed by leptin receptor knockdown. Silencing TGF-β in MSCs also abolished OS cell chemoresistance induced by leptin-conditioned medium. Cisplatin resistance was also induced when leptin-conditioned MSCs were co-injected with MG-63 OS cells to generate subcutaneous xenografts in nude mice. Finally, we observed a significant correlation between autophagy-associated gene expression in OS clinical samples and patient prognosis. We conclude that leptin upregulates TGF-β in MSCs, which promotes autophagy-mediated chemoresistance in OS cells.
Collapse
Affiliation(s)
- Helin Feng
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
- Postdoctoral Mobile Station, Hebei Medical University, Shijiazhuang 050017, Hebei, China
- Hebei Province Xingtai People’s Hospital Postdoctoral Workstation, Xingtai 054031, Hebei, China
| | - Qianqian Zhang
- Department of Gynecology, Hebei Medical University Second Affiliated Hospital, Shijiazhuang 050000, Hebei, China
| | - Yi Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Lili Zhao
- Hebei Province Xingtai People’s Hospital Postdoctoral Workstation, Xingtai 054031, Hebei, China
| | - Baoen Shan
- Postdoctoral Mobile Station, Hebei Medical University, Shijiazhuang 050017, Hebei, China
- Research Centre, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| |
Collapse
|
13
|
Li J, Gao Y, Yu T, Lange JK, LeBoff MS, Gorska A, Luu S, Zhou S, Glowacki J. Obesity and leptin influence vitamin D metabolism and action in human marrow stromal cells. J Steroid Biochem Mol Biol 2020; 198:105564. [PMID: 31809868 DOI: 10.1016/j.jsbmb.2019.105564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 02/05/2023]
Abstract
Obesity is associated with low serum 25-hydroxyvitamin D [s25(OH)D], high serum leptin, and generally high bone mineral density (BMD). Human Marrow Stromal Cells (hMSCs) differentiate to osteoblasts and are both a target and source of vitamin D metabolites in bone marrow. There is no information about the influence of obesity on vitamin D metabolism and osteoblastogenesis in hMSCs and little about direct effects of leptin on hMSCs. In this study, we tested the hypotheses that 1) obesity has an influence on the ex vivo constitutive expression of vitamin D-hydroxylase genes in hMSCs, and 2) recombinant human (rh) Leptin regulates the D-hydroxylases and promotes osteoblastogenesis in hMSCs. In a cohort of female subjects undergoing joint replacement surgery, the effects of Body Mass Index (BMI) and Fat Mass Index (FMI) on BMD T-scores and s25(OH)D were evaluated. hMSCs were isolated from bone tissues discarded during surgery. The direct effects of rh-Leptin on osteoblast differentiation and D-related genes in hMSCs were examined in vitro. There were positive correlations for BMD T-score of femoral neck and spine with BMI and FMI. Serum 25(OH)D levels in obese subjects were 71% of that in non-obese counterparts (p = 0.001). hMSCs from obese women had higher constitutive expression of CYP27A1/25-hydroxylase and vitamin D receptor. Those findings raised the mechanistic question of how obesity could influence vitamin D metabolism and osteoblast differentiation in hMSCs. Treating hMSCs with rh-Leptin in vitro significantly stimulated osteoblastogenesis. In addition, leptin downregulated CYP24A1 and upregulated CYP27B1, CYP27A1 and VDR, which play vital roles in vitamin D metabolism. Furthermore, co-treatment with leptin and vitamin D3 metabolites promoted ALP activity compared with either alone. This research demonstrates links between obesity, vitamin D metabolism, and osteoblastogenesis by which leptin's direct effects on D-metabolism and osteoblast differentiation in hMSCs may protect bone from low s25(OH)D in obese subjects.
Collapse
Affiliation(s)
- Jing Li
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Endocrinology, West China Hospital, Sichuan University West China School of Medicine, Chengdu, Sichuan, China
| | - Yuan Gao
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Yu
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jeffrey K Lange
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Meryl S LeBoff
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anna Gorska
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Luu
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Julie Glowacki
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Oral & Maxillofacial Surgery, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
14
|
Idelevich A, Sato K, Nagano K, Rowe G, Gori F, Baron R. ΔFosB Requires Galanin, but not Leptin, to Increase Bone Mass via the Hypothalamus, but both are needed to increase Energy expenditure. J Bone Miner Res 2019; 34:1707-1720. [PMID: 30998833 PMCID: PMC6744351 DOI: 10.1002/jbmr.3741] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/22/2019] [Accepted: 04/05/2019] [Indexed: 01/29/2023]
Abstract
Energy metabolism and bone homeostasis share several regulatory pathways. The AP1 transcription factor ΔFosB and leptin both regulate energy metabolism and bone, yet whether their pathways intersect is not known. Transgenic mice overexpressing ΔFosB under the control of the Enolase 2 (ENO2) promoter exhibit high bone mass, high energy expenditure, low fat mass, and low circulating leptin levels. Because leptin is a regulator of bone and ΔFosB acts on leptin-responsive ventral hypothalamic (VHT) neurons to induce bone anabolism, we hypothesized that regulation of leptin may contribute to the central actions of ΔFosB in the VHT. To address this question, we used adeno-associated virus (AAV) expression of ΔFosB in the VHT of leptin-deficient ob/ob mice and genetic crossing of ENO2-ΔFosB with ob/ob mice. In both models, leptin deficiency prevented ΔFosB-triggered reduction in body weight, increase in energy expenditure, increase in glucose utilization, and reduction in pancreatic islet size. In contrast, leptin deficiency failed to prevent ΔFosB-triggered increase in bone mass. Unlike leptin deficiency, galanin deficiency blocked both the metabolic and the bone ΔFosB-induced effects. Overall, our data demonstrate that, while the catabolic energy metabolism effects of ΔFosB require intact leptin and galanin signaling, the bone mass-accruing effects of ΔFosB require galanin but are independent of leptin. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Anna Idelevich
- Department of Medicine, Harvard Medical School and Endocrine Unit, Massachusetts General Hospital, and Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Kazusa Sato
- Department of Medicine, Harvard Medical School and Endocrine Unit, Massachusetts General Hospital, and Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Kenichi Nagano
- Department of Medicine, Harvard Medical School and Endocrine Unit, Massachusetts General Hospital, and Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Glenn Rowe
- Department of Medicine, Harvard Medical School and Endocrine Unit, Massachusetts General Hospital, and Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Francesca Gori
- Department of Medicine, Harvard Medical School and Endocrine Unit, Massachusetts General Hospital, and Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Roland Baron
- Department of Medicine, Harvard Medical School and Endocrine Unit, Massachusetts General Hospital, and Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
15
|
The Analysis of In Vivo Aging in Human Bone Marrow Mesenchymal Stromal Cells Using Colony-Forming Unit-Fibroblast Assay and the CD45 lowCD271 + Phenotype. Stem Cells Int 2019; 2019:5197983. [PMID: 31467563 PMCID: PMC6701348 DOI: 10.1155/2019/5197983] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/28/2019] [Accepted: 07/14/2019] [Indexed: 12/13/2022] Open
Abstract
Uncultured mesenchymal stromal cells (MSCs) are increasingly used in therapies; however, the effects of donor age on their biological characteristics and gene expression remain unclear. The aim of this study was to investigate age-related changes in bone marrow (BM) MSCs following minimal or no culture manipulation. Iliac crest BM was aspirated from 67 healthy donors (19-89 years old) and directly used for the colony-forming unit-fibroblast (CFU-F) assay or CD45lowCD271+ cell enumeration. The colonies were analysed for colony area and integrated density (ID) when grown in standard MSC media or media supplemented with human serum from young (YS) or old (OS) donors. There was a notable age-related decline in the number of MSCs per millilitre of BM aspirate revealed by the CFU-F assay (r = −0.527, p < 0.0001) or flow cytometry (r = −0.307, p = 0.0116). Compared to young donors (19-40 years old), colony IDs were significantly lower in older donors (61-89 years old), particularly for smaller-sized colonies (42% lower, p < 0.01). When cultured in media supplemented with OS, young and old donor MSCs formed colonies with lower IDs, by 21%, p < 0.0001, and 27%, p < 0.05, respectively, indicating the formation of smaller sparser colonies. No significant differences in the expression of selected adipogenic, osteogenic, stromal, and bone remodelling genes as well as CD295, CD146, CD106, and connexin 43 surface molecules were found in sorted CD45lowCD271+ MSCs from young and old donors (n = 8 donors each). Altogether, these results show similar trends for age-related decline in BM MSC numbers measured by the CFU-F assay and flow cytometry and reveal age-related effects of human serum on MSC colony formation. No significant differences in selected gene expression in uncultured CD45lowCD271+ MSCs suggest that old donor MSCs may not be inferior in regard to their multipotential functions. Due to large donor-to-donor variation in all donor groups, our data indicate that an individual's chronological age is not a reliable predictor of their MSC number or potency.
Collapse
|
16
|
Liu X, Li Z, Liu H, Zhu Y, Xia D, Wang S, Gu R, Wu W, Zhang P, Liu Y, Zhou Y. Low concentration flufenamic acid enhances osteogenic differentiation of mesenchymal stem cells and suppresses bone loss by inhibition of the NF-κB signaling pathway. Stem Cell Res Ther 2019; 10:213. [PMID: 31324207 PMCID: PMC6642517 DOI: 10.1186/s13287-019-1321-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/09/2019] [Accepted: 06/30/2019] [Indexed: 02/02/2023] Open
Abstract
Background As the representative of fenamic acids, an important group of NSAIDs, flufenamic acid (FFA) has been used for anti-inflammation and analgesia in the clinic. Recently, researches have focused on the role of some members of NSAIDs in promoting osteogenesis. However, little attention has been paid to the subgroup of fenamic acids, and it remains unclear whether FFA and other fenamic acids could regulate mesenchymal stem cells’ (MSCs) lineage commitment and bone regeneration. Methods Here we treated two kinds of human MSCs with FFA at different concentrations in vitro and examined the effect of FFA on osteogenic differentiation of human MSCs. This was followed by heterotopic bone formation assay in nude mice. In addition, ovariectomized and aged mice were used as osteoporotic models to test the effect of FFA on osteoporosis. Besides, activators and inhibitor of nuclear factor-κB (NF-κB) signaling pathway and western blot were used to clarify the mechanism of the promoting effect of low concentration FFA on osteogenesis. Results Our results indicated that low concentrations of FFA could significantly enhance osteogenic differentiation of human MSCs in vitro, as well as in vivo. In addition, FFA treatment suppressed bone loss in ovariectomized and aged mice. Mechanistically, FFA at low concentrations promoted osteogenesis differentiation of human MSCs by inhibition of the NF-κB signaling pathway. Conclusions Collectively, our study suggested that low concentration FFA could be used in bone tissue engineering or osteoporosis by promoting osteogenic differentiation of human MSCs. Electronic supplementary material The online version of this article (10.1186/s13287-019-1321-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Hao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Dandan Xia
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, 100871, People's Republic of China
| | - Siyi Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Weiliang Wu
- Department of Implantology II, The Affiliated Stomatological Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| |
Collapse
|
17
|
Reid IR, Baldock PA, Cornish J. Effects of Leptin on the Skeleton. Endocr Rev 2018; 39:938-959. [PMID: 30184053 DOI: 10.1210/er.2017-00226] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 06/26/2018] [Indexed: 12/12/2022]
Abstract
Leptin originates in adipocytes, including those in bone marrow, and circulates in concentrations 20 to 90 times higher than those in the cerebrospinal fluid. It has direct anabolic effects on osteoblasts and chondrocytes, but it also influences bone indirectly, via the hypothalamus and sympathetic nervous system, via changes in body weight, and via effects on the production of other hormones (e.g., pituitary). Leptin's role in bone physiology is determined by the balance of these conflicting effects. Reflecting this inconsistency, the leptin-deficient mouse has reduced length and bone mineral content of long bones but increased vertebral trabecular bone. A consistent bone phenotype in human leptin deficiency has not been established. Systemic leptin administration in animals and humans usually exerts a positive effect on bone mass, and leptin administration into the cerebral ventricles usually normalizes the bone phenotype in leptin-deficient mice. Reflecting the role of the sympathetic nervous system in mediating the central catabolic effects of leptin on the skeleton, β-adrenergic agonists and antagonists have major effects on bone in mice, but this is not consistently seen in humans. The balance of the central and peripheral effects of leptin on bone remains an area of substantial controversy and might vary between species and according to other factors such as body weight, baseline circulating leptin levels, and the presence of specific pathologies. In humans, leptin is likely to contribute to the positive relationship observed between adiposity and bone density, which allows the skeleton to respond appropriately to changes in soft tissue mass.
Collapse
Affiliation(s)
- Ian R Reid
- University of Auckland, Auckland, New Zealand.,Department of Endocrinology, Auckland District Health Board, Auckland, New Zealand
| | - Paul A Baldock
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | |
Collapse
|
18
|
Chen S, Zhu H, Wang G, Xie Z, Wang J, Chen J. Combined use of leptin and mechanical stress has osteogenic effects on ossification of the posterior longitudinal ligament. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2018; 27:1757-1766. [PMID: 29909551 DOI: 10.1007/s00586-018-5663-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/04/2018] [Accepted: 06/06/2018] [Indexed: 11/26/2022]
Abstract
PURPOSE To evaluate the effects of leptin/leptin receptor (LepR) combined with mechanical stress on the development of ossification of the posterior longitudinal ligament (OPLL), which is a disease characterized by ectopic bone formation of the posterior longitudinal ligament (PLL) and can lead to radiculopathy and myelopathy. METHODS Six human samples of the PLL were analyzed for the expression of leptin and LepR by RT-PCR and western blotting. PLL cells were stimulated with leptin and mechanical stress delivered via a Flexcell tension system, and osteogenic differentiation was evaluated by RT-PCR and western blotting analysis of osteogenic marker expression as well as by alkaline phosphatase (ALP) staining and alizarin red S staining. Activation of mitogen-activated protein kinase (MAPK), Janus kinase (JAK) 2-signal transducer, activator of transcription (STAT) 3 and phosphatidylinositol 3-kinase (PI3K)-Akt was evaluated by western blotting. RESULTS Samples from the OPLL group had higher LepR mRNA and protein levels and lower leptin levels than those from healthy controls. Exposure to leptin and Flexcell increased the number of ALP-positive cells and calcium nodules in a dose-dependent manner; this effect was accompanied by upregulation of the osteogenic markers osteocalcin, runt-related transcription factor 2 (RUNX2) and osteopontin. Extracellular signal-regulated kinase, P38 MAPK, JAK2, STAT3, PI3K and Akt signaling, was also activated by the combined effects of leptin and mechanical stress. CONCLUSIONS Leptin and LepR are differentially expressed in OPLL tissues, and the combined use of leptin/LepR and mechanical stress promotes osteogenic differentiation of PLL cells via MAPK, JAK2-STAT3 and PI3K/Akt signaling. These slides can be retrieved under Electronic Supplementary Material.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310020, China
- Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Haifeng Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310020, China
- Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Gangliang Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310020, China
- Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310020, China
- Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Jiying Wang
- Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310020, China.
- Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
19
|
Li J, Zuo B, Zhang L, Dai L, Zhang X. Osteoblast versus Adipocyte: Bone Marrow Microenvironment-Guided Epigenetic Control. CASE REPORTS IN ORTHOPEDIC RESEARCH 2018. [DOI: 10.1159/000489053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The commitment and differentiation of bone marrow mesenchymal stem cells (MSCs) is tightly controlled by the local environment ensuring lineage differentiation balance and bone homeostasis. However, pathological conditions linked with osteoporosis have changed the bone marrow microenvironment, shifting MSCs’ fate to favor adipocytes over osteoblasts, and consequently leading to decreased bone mass with marrow fat accumulation. Multiple questions related to the underlying mechanisms remain to be answered. As recent findings have confirmed the fundamental role of the epigenetic mechanism in connecting environmental signals with gene expression and stem cell differentiation, a regulatory network in the bone marrow microenvironment, epigenetic modulation, gene expression, and MSC differentiation begins to emerge. This review discusses how pathological environmental factors affect MSCs’ fate by epigenetic modulating lineage-specific genes. We conclude that manipulating local environments and/or the epigenetic regulatory machinery that target the adipocyte differentiation pathway might be a therapeutic implication of bone loss diseases such as osteoporosis.
Collapse
|
20
|
Luciani P, Fibbi B, Mazzanti B, Deledda C, Ballerini L, Aldinucci A, Benvenuti S, Saccardi R, Peri A. The effects of Exendin-4 on bone marrow-derived mesenchymal cells. Endocrine 2018; 60:423-434. [PMID: 29094257 DOI: 10.1007/s12020-017-1430-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/16/2017] [Indexed: 12/14/2022]
Abstract
PURPOSE GLP-1 receptor agonists are antidiabetic drugs currently used in the therapy of type 2 diabetes. Despite several in vitro and in vivo animal studies suggesting a beneficial effect of GLP-1 analogues on bone, in humans their skeletal effects are not clear and clinical studies report conflicting results. METHODS We differentiated human mesenchymal stromal cells (hMSC) toward the adipogenic and the osteoblastic lineages, analysing the effect of Exendin-4 (EXE) before, during and after specific differentiations. RESULTS We showed EXE ability to act selectively on a sub-population of hMSC characterised by a more stem potential, shifting them from G1 to S/M phase of cell cycle. We observed that EXE pre-treatment promotes both adipogenic and osteoblastic differentiations, possibly determined by an increased number of uncommitted progenitors. In fully differentiated cells, EXE affects mature adipocytes by increasing lipolysis, otherwise not altering osteoblasts metabolic activity. Moreover, the increased expression of osteoprotegerin, a modulator of the RANK/RANKL system, observed during osteogenic induction in presence of EXE, could negatively modulate osteoclastogenesis. CONCLUSIONS Our data suggest a complex action of EXE on bone, targeting the proliferation of mesenchymal progenitors, the metabolism of mature adipocytes and the modulation of osteoclastogenesis. Thus, an overall positive effect of this molecule on bone quality might be hypothesised.
Collapse
Affiliation(s)
- Paola Luciani
- Endocrine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Benedetta Fibbi
- Endocrine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Benedetta Mazzanti
- Haematology Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cristiana Deledda
- Endocrine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lara Ballerini
- Haematology Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Susanna Benvenuti
- Endocrine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Riccardo Saccardi
- Haematology Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro Peri
- Endocrine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| |
Collapse
|
21
|
Vajapeyam S, Ecklund K, Mulkern RV, Feldman HA, O'Donnell JM, DiVasta AD, Rosen CJ, Gordon CM. Magnetic resonance imaging and spectroscopy evidence of efficacy for adrenal and gonadal hormone replacement therapy in anorexia nervosa. Bone 2018; 110:335-342. [PMID: 29496516 PMCID: PMC5879439 DOI: 10.1016/j.bone.2018.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/19/2018] [Accepted: 02/21/2018] [Indexed: 10/17/2022]
Abstract
PURPOSE Dehydroepiandrosterone (DHEA)+estrogen/progestin therapy for adolescent girls with anorexia nervosa (AN) has the potential to arrest bone loss. The primary aim of this study was to test the effects of DHEA+estrogen/progestin therapy in adolescent girls with AN on bone marrow in the distal femur using magnetic resonance imaging (MRI) and spectroscopy. METHODS Seventy adolescent girls with AN were enrolled in a double blind, randomized, placebo-controlled trial at two urban hospital-based programs. INTERVENTION Seventy-six girls were randomly assigned to receive 12months of either oral micronized DHEA or placebo. DHEA was administered with conjugated equine estrogens (0.3mg daily) for 3months, then an oral contraceptive (20μg ethinyl estradiol/ 0.1mg levonorgestrel) for 9months. The primary outcome measure was bone marrow fat by MRI and magnetic resonance spectroscopy (MRS). RESULTS T2 of the water resonance dropped significantly less in the active vs. placebo group over 12months at both the medial and lateral distal femur (p=0.02). Body mass index (BMI) was a significant effect modifier for T1 and for T2 of unsaturated (T2unsat) and saturated fat (T2sat) in the lateral distal femur. Positive effects of the treatment of DHEA+estrogen/progestin were seen primarily for girls above a BMI of about 18kg/m2. CONCLUSIONS These findings suggest treatment with oral DHEA+estrogen/progestin arrests the age- and disease-related changes in marrow fat composition in the lateral distal femur reported previously in this population.
Collapse
Affiliation(s)
- Sridhar Vajapeyam
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Kirsten Ecklund
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Robert V Mulkern
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Henry A Feldman
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, USA
| | | | - Amy D DiVasta
- Division of Adolescent/Young Adult Medicine, Boston Children's Hospital, Boston, MA, USA
| | | | - Catherine M Gordon
- Division of Adolescent and Transition Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
22
|
Fallahnezhad S, Piryaei A, Darbandi H, Amini A, Ghoreishi SK, Jalalifirouzkouhi R, Bayat M. Effect of low‐level laser therapy and oxytocin on osteoporotic bone marrow‐derived mesenchymal stem cells. J Cell Biochem 2018; 119:983-997. [DOI: 10.1002/jcb.26265] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022]
Abstract
Postmenopausal osteoporosis (OP) is a major concern for public health. Low‐level laser therapy (LLLT) has a positive effect on the health of bone marrow mesenchymal stem cells (BMMSCs). The purpose of this study is to evaluate the influence of LLLT and oxytocin (OT) incubation—individually and in combination—on osteoporotic BMMSCs in ovariectomized rats. Twelve female rats were randomized into two groups to undergo either a sham surgery (sham group) or ovariectomy‐induced osteoporosis (OVX group). MSCs harvested from the BM of healthy and OVX rats underwent culture expansion. There were five groups. In Groups one (sham‐BMMSC) and two (OVX‐BMMSC) the cells were held in osteogenic condition medium without any intervention. In the group three (OT), OT incubation with optimum dose was performed for 48 h (two times, 10−12 molar). In Group four, laser‐treated‐OVX‐BMMSCs were treated with optimum protocol of LLLT (one time, 1.2 J/cm2). In Group five (laser + OT group), the OT incubation plus the laser irradiation was performed. The biostimulatory effect of LLLT is demonstrated by a significant increase in the viability of OVX‐BMMSCs, cell cycle, and extracellular levels of Transforming growth factor beta (TGF‐β), insulin‐like growth factor‐I (IGF‐I), and Alkaline phosphatase (ALP) compared to control OVX‐BMMSCs and/or the sham group. OT incubation and laser + OT incubation have a positive effect on OVX‐BMMSCs. However, LLLT is more effective statistically. We conclude that LLLT significantly improved cell viability, enhanced the osteogenic potential of the OVX‐BMMSCs, and increased the extracellular levels of the TGF‐β, IGF‐I, and ALP.
Collapse
Affiliation(s)
- Somaye Fallahnezhad
- Department of Biology and Anatomical Sciences School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Hasan Darbandi
- Department of Immunology School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| | | | | | - Mohammad Bayat
- Cellular and Molecular Biology Research Center, and Department of Biology and Anatomical Sciences School of Medicine Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
23
|
Huang H, Cheng WX, Hu YP, Chen JH, Zheng ZT, Zhang P. Relationship between heterotopic ossification and traumatic brain injury: Why severe traumatic brain injury increases the risk of heterotopic ossification. J Orthop Translat 2017; 12:16-25. [PMID: 29662775 PMCID: PMC5866497 DOI: 10.1016/j.jot.2017.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/12/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023] Open
Abstract
Heterotopic ossification (HO) is a pathological phenomenon in which ectopic lamellar bone forms in soft tissues. HO involves many predisposing factors, including congenital and postnatal factors. Postnatal HO is usually induced by fracture, burn, neurological damage (brain injury and spinal cord injury) and joint replacement. Recent studies have found that patients who suffered from bone fracture combined with severe traumatic brain injury (S-TBI) are at a significantly increased risk for HO occurrence. Thus, considerable research focused on the influence of S-TBI on fracture healing and bone formation, as well as on the changes in various osteogenic factors with S-TBI occurrence. Brain damage promotes bone formation, but the exact mechanisms underlying bone formation and HO after S-TBI remain to be clarified. Hence, this article summarises the findings of previous studies on the relationship between S-TBI and HO and discusses the probable causes and mechanisms of HO caused by S-TBI. The translational potential of this article: A better understanding of the probable causes of traumatic brain injury-induced HO can provide new perspectives and ideas in preventing HO and may support to design more targeted therapies to reduce HO or enhance the bone formation.
Collapse
Affiliation(s)
- Huan Huang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wen-Xiang Cheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Ping Hu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian-Hai Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zheng-Tan Zheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Peng Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
24
|
Ecklund K, Vajapeyam S, Mulkern RV, Feldman HA, O'Donnell JM, DiVasta AD, Gordon CM. Bone marrow fat content in 70 adolescent girls with anorexia nervosa: Magnetic resonance imaging and magnetic resonance spectroscopy assessment. Pediatr Radiol 2017; 47:952-962. [PMID: 28432403 PMCID: PMC5650065 DOI: 10.1007/s00247-017-3856-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/14/2017] [Accepted: 03/30/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Adolescents and women with anorexia nervosa have increased bone marrow fat and decreased bone formation, at least in part due to hormonal changes leading to preferential stem cell differentiation to adipocytes over osteoblasts. OBJECTIVE The purpose of this study was to evaluate marrow fat content and correlate with age and disease severity using knee MRI with T1 relaxometry (T1-R) and MR spectroscopy (MRS) in 70 adolescents with anorexia nervosa. MATERIALS AND METHODS We enrolled 70 girls with anorexia nervosa who underwent 3-T knee MRI with coronal T1-W images, T1-R and single-voxel proton MRS at 30 and 60 ms TE. Metaphyses were scored visually on the T1-W images for red marrow. Visual T1 score, T1 relaxometry values, MRS lipid indices and fat fractions were analyzed by regression on age, body mass index (BMI) and bone mineral density (BMD) as disease severity markers. MRS measures included unsaturated fat index, T2 water, unsaturated and saturated fat fractions. RESULTS All red marrow measures declined significantly with age. T1-R values were associated negatively with BMI and BMD for girls ≤16 years (P=0.03 and P=0.002, respectively) and positively for those≥17 years (P=0.05 and P=0.003, respectively). MRS identified a strong inverse association between T2 water and saturated fat fraction from 60 ms TE data (r=-0.85, P<0.0001). There was no association between unsaturated fat index and BMI or BMD. CONCLUSIONS The association between T1 and BMI and BMD among older girls suggests more marrow fat in those with severe anorexia nervosa. In contrast, the physiological association between marrow fat content and age remained dominant in younger patients. The strong association between T2 water and saturated fat may relate to the restricted mobility of water with increasing marrow fat.
Collapse
Affiliation(s)
- Kirsten Ecklund
- Department of Radiology, Boston Children's Hospital, 333 Longwood Ave., Boston, MA, 02115, USA.
| | - Sridhar Vajapeyam
- Department of Radiology, Boston Children's Hospital, 333 Longwood Ave., Boston, MA, 02115, USA
| | - Robert V Mulkern
- Department of Radiology, Boston Children's Hospital, 333 Longwood Ave., Boston, MA, 02115, USA
| | - Henry A Feldman
- Clinical Research Center, Boston Children's Hospital, Boston, MA, USA
| | | | - Amy D DiVasta
- Division of Adolescent/Young Adult Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Catherine M Gordon
- Division of Adolescent and Transition Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
25
|
Zheng B, Jiang J, Chen Y, Lin M, Du Z, Xiao Y, Luo K, Yan F. Leptin Overexpression in Bone Marrow Stromal Cells Promotes Periodontal Regeneration in a Rat Model of Osteoporosis. J Periodontol 2017; 88:808-818. [PMID: 28440742 DOI: 10.1902/jop.2017.170042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Osteoporosis is associated with widespread periodontitis and impaired periodontal healing. However, there is a lack of information about the outcomes of regenerative approaches under the influence of osteoporosis. This study investigates the effect of leptin (LEP) overexpression on the regenerative potential of bone marrow stromal cells (BMSCs) in an osteoporotic rat periodontal fenestration defect model. METHODS Rat BMSCs were transfected with adenoviruses harboring the human (h)LEP gene. Cell proliferation and osteogenic differentiation were evaluated. A β-tricalcium phosphate scaffold seeded with transfected cells was implanted into nude mice to investigate ectopic osteogenesis and into an osteoporotic rat defect to study periodontal regeneration. Regenerated periodontal and bone-like tissues were analyzed by histologic methods. RESULTS hLEP overexpression induced osteogenic differentiation of BMSCs as evidenced by the upregulation of osteogenesis-related genes such as Runt-related transcription factor 2, alkaline phosphatase (ALP), and collagen Type I, as well as increased ALP activity and enhanced mineralization. Mice implanted with hLEP-BMSC-containing scaffolds showed more extensive formation of bone-like tissue than those in other groups. Periodontal defects were also filled to a greater degree when treated with hLEP-BMSCs and contained cementum and a well-organized periodontal ligament after 10 and 28 days. CONCLUSION hLEP overexpression in BMSCs can stimulate periodontal regeneration in osteoporotic conditions and might be a promising strategy for periodontal regeneration in patients with osteoporosis.
Collapse
Affiliation(s)
- Baoyu Zheng
- Department of Periodontology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jun Jiang
- Fujian Biological Materials Engineering and Technology Center of Stomatology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yuling Chen
- Fujian Biological Materials Engineering and Technology Center of Stomatology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Minkui Lin
- Fujian Biological Materials Engineering and Technology Center of Stomatology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhibin Du
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kai Luo
- Fujian Biological Materials Engineering and Technology Center of Stomatology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Fuhua Yan
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
26
|
Hannema SE, Wit JM, Houdijk MECAM, van Haeringen A, Bik EC, Verkerk AJMH, Uitterlinden AG, Kant SG, Oostdijk W, Bakker E, Delemarre-van de Waal HA, Losekoot M. Novel Leptin Receptor Mutations Identified in Two Girls with Severe Obesity Are Associated with Increased Bone Mineral Density. Horm Res Paediatr 2017; 85:412-20. [PMID: 26925581 DOI: 10.1159/000444055] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/14/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recessive mutations in the leptin receptor (LEPR) are a rare cause of hyperphagia and severe early-onset obesity. To date, the phenotype has only been described in 25 obese children, some of whom also had altered immune function, hypogonadotropic hypogonadism, reduced growth hormone secretion, hypothalamic hypothyroidism or reduced adult height. We provide a detailed description of the phenotype of 2 affected girls to add to this knowledge. METHODS Whole-exome sequencing and targeted sequencing were used to detect the LEPR mutations. RNA analysis was performed to assess the effect of splice-site mutations. RESULTS In 2 unrelated girls with severe obesity, three novel LEPR mutations were detected. Longitudinal growth data show normal childhood growth, and in the older girl, a normal adult height despite hypogonadotropic hypogonadism and the lack of an obvious pubertal growth spurt. Bone age is remarkably advanced in the younger (prepubertal) girl, and bone mineral density (BMD) is high in both girls, which might be directly or indirectly related to leptin resistance. CONCLUSION The spectrum of clinical features of LEPR deficiency may be expanded with increased BMD. Future observations in LEPR-deficient subjects should help further unravel the role of leptin in human bone biology.
Collapse
Affiliation(s)
- Sabine E Hannema
- Department of Paediatrics, Willem Alexander Children's Hospital, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Jin C, Zhang P, Zhang M, Zhang X, Lv L, Liu H, Liu Y, Zhou Y. Inhibition of SLC7A11 by Sulfasalazine Enhances Osteogenic Differentiation of Mesenchymal Stem Cells by Modulating BMP2/4 Expression and Suppresses Bone Loss in Ovariectomized Mice. J Bone Miner Res 2017; 32:508-521. [PMID: 27696501 DOI: 10.1002/jbmr.3009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/24/2016] [Accepted: 09/29/2016] [Indexed: 12/27/2022]
Abstract
An imbalance in osteogenesis and adipogenesis is a crucial pathological factor in the development of osteoporosis. Many attempts have been made to develop drugs to prevent and treat this disease. In the present study, we investigated the phenomenon whereby downregulation of SLC7A11 significantly enhanced the osteogenic differentiation of mesenchymal stem cells (MSCs) in vitro, and promoted the bone formation in vivo. Sulfasalazine (SAS), an inhibitor of SLC7A11, increased the osteogenic potential effectively. Mechanistically, inhibition of SLC7A11 by SAS treatment or knockdown of SLC7A11 increased BMP2/4 expression dramatically. In addition, we detected increased Slc7a11 expression in bone marrow MSCs of ovariectomized (OVX) mice. Remarkably, SAS treatment attenuated bone loss in ovariectomized mice. Together, our data suggested that SAS could be used to treat osteoporosis by enhancing osteogenic differentiation of MSCs. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Chanyuan Jin
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Min Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
28
|
Hamrick MW. Role of the Cytokine-like Hormone Leptin in Muscle-bone Crosstalk with Aging. J Bone Metab 2017; 24:1-8. [PMID: 28326295 PMCID: PMC5357607 DOI: 10.11005/jbm.2017.24.1.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022] Open
Abstract
The cytokine-like hormone leptin is a classic adipokine that is secreted by adipocytes, increases with weight gain, and decreases with weight loss. Additional studies have, however, shown that leptin is also produced by skeletal muscle, and leptin receptors are abundant in both skeletal muscle and bone-derived mesenchymal (stromal) stem cells. These findings suggest that leptin may play an important role in muscle-bone crosstalk. Leptin treatment in vitro increases the expression of myogenic genes in primary myoblasts, and leptin treatment in vivo increases the expression of microRNAs involved in myogenesis. Bone marrow adipogenesis is associated with low bone mass in humans and rodents, and leptin can reduce marrow adipogenesis centrally through its receptors in the hypothalamus as well as directly via its receptors in bone marrow stem cells. Yet, central leptin resistance can increase with age, and low circulating levels of leptin have been observed among the frail elderly. Thus, aging appears to significantly alter leptin-mediated crosstalk among various organs and tissues. Aging is associated with bone loss and muscle atrophy, contributing to frailty, postural instability, and the incidence of falls. Therapeutic interventions such as protein and amino acid supplementation that can increase muscle mass and muscle-derived leptin may have multiple benefits for the elderly that can potentially reduce the incidence of falls and fractures.
Collapse
Affiliation(s)
- Mark W. Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
29
|
Shi Y, Du L, Lin L, Wang Y. Tumour-associated mesenchymal stem/stromal cells: emerging therapeutic targets. Nat Rev Drug Discov 2016; 16:35-52. [PMID: 27811929 DOI: 10.1038/nrd.2016.193] [Citation(s) in RCA: 342] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells, also known as mesenchymal stromal cells (MSCs), exist in many tissues and are known to actively migrate to sites of tissue injury, where they participate in wound repair. Tumours can be considered "wounds that never heal" and, in response to cues from a tumour, MSCs are continuously recruited to and become integral components of the tumour microenvironment. Recently, it has become apparent that such tumour-associated MSCs (TA-MSCs) have an active role in tumour initiation, promotion, progression and metastasis. In this Review, we discuss recent advances in our understanding of the pathogenic role of TA-MSCs in regulating the survival, proliferation, migration and drug resistance of tumour cells, as well as the influence of MSCs on the immune status of the tumour microenvironment. Moreover, we discuss therapeutic approaches that target TA-MSC upstream or downstream modulators or use MSCs as vehicles for the delivery of tumoricidal agents. It is anticipated that new insights into the functions of TA-MSCs will lead to the development of novel therapeutic strategies against tumours.
Collapse
Affiliation(s)
- Yufang Shi
- The First Affiliated Hospital of Soochow University and Jiangsu Engineering Research Center for Tumor Immunotherapy, Institutes for Translational Medicine and Suzhou Key Laboratory of Tumor Microenvironment and Pathology, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China.,Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901, USA.,Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Liming Du
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Liangyu Lin
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China.,Shanghai Jiao Tong University School of Medicine, 280 Chongqing Road, Shanghai 200025, China
| | - Ying Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| |
Collapse
|
30
|
Román F, Urra C, Porras O, Pino AM, Rosen CJ, Rodríguez JP. Real-Time H 2 O 2 Measurements in Bone Marrow Mesenchymal Stem Cells (MSCs) Show Increased Antioxidant Capacity in Cells From Osteoporotic Women. J Cell Biochem 2016; 118:585-593. [PMID: 27632788 DOI: 10.1002/jcb.25739] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 01/21/2023]
Abstract
Oxidative stress (OS) derived from an increase in intracellular reactive oxygen species (ROS) is a major determinant of aging and lifespan. It has also been associated with several age-related disorders, like postmenopausal osteoporosis of Mesenchymal stem cells (MSCs). MSCs are the common precursors for osteoblasts and adipocytes; appropriate commitment and differentiation of MSCs into a specific phenotype is modulated, among other factors, by ROS balance. MSCs have shown more resistance to ROS than differentiated cells, and their redox status depends on complex and abundant anti-oxidant mechanisms. The purpose of this work was to analyze in real time, H2 O2 signaling in individual h-MSCs, and to compare the kinetic parameters of H2 O2 management by cells derived from both control (c-) and osteoporotic (o-) women. For these purposes, cells were infected with a genetically encoded fluorescent biosensor named HyPer, which is specific for detecting H2 O2 inside living cells. Subsequently, cells were sequentially challenged with 50 and 500 μM H2 O2 pulses, and the cellular response was recorded in real time. The results demonstrated adequate expression of the biosensor allowing registering fluorescence from HyPer at a single cell level. Comparison of the response of c- and o-MSCs to the oxidant challenges demonstrated improved antioxidant activity in o-MSCs. This was further corroborated by measuring the relative expression of mRNAs for catalase, superoxide dismutase-1, thioredoxine, and peroxiredoxine, as well as by cell-surviving capacity under short-term H2 O2 treatment. We conclude that functional differences exist between healthy and osteoporotic human MSCs. The mechanism for these differences requires further study. J. Cell. Biochem. 118: 585-593, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Flavia Román
- Laboratorio de Biología Celular, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Carla Urra
- Laboratorio de Biología Celular, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Omar Porras
- Laboratorio de Biología Celular, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Ana María Pino
- Laboratorio de Biología Celular, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | | | - Juan Pablo Rodríguez
- Laboratorio de Biología Celular, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| |
Collapse
|
31
|
Iwaniec UT, Turner RT. Influence of body weight on bone mass, architecture and turnover. J Endocrinol 2016; 230:R115-30. [PMID: 27352896 PMCID: PMC4980254 DOI: 10.1530/joe-16-0089] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
Weight-dependent loading of the skeleton plays an important role in establishing and maintaining bone mass and strength. This review focuses on mechanical signaling induced by body weight as an essential mechanism for maintaining bone health. In addition, the skeletal effects of deviation from normal weight are discussed. The magnitude of mechanical strain experienced by bone during normal activities is remarkably similar among vertebrates, regardless of size, supporting the existence of a conserved regulatory mechanism, or mechanostat, that senses mechanical strain. The mechanostat functions as an adaptive mechanism to optimize bone mass and architecture based on prevailing mechanical strain. Changes in weight, due to altered mass, weightlessness (spaceflight), and hypergravity (modeled by centrifugation), induce an adaptive skeletal response. However, the precise mechanisms governing the skeletal response are incompletely understood. Furthermore, establishing whether the adaptive response maintains the mechanical competence of the skeleton has proven difficult, necessitating the development of surrogate measures of bone quality. The mechanostat is influenced by regulatory inputs to facilitate non-mechanical functions of the skeleton, such as mineral homeostasis, as well as hormones and energy/nutrient availability that support bone metabolism. Although the skeleton is very capable of adapting to changes in weight, the mechanostat has limits. At the limits, extreme deviations from normal weight and body composition are associated with impaired optimization of bone strength to prevailing body size.
Collapse
Affiliation(s)
- Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| | - Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| |
Collapse
|
32
|
Li J, Liu X, Zuo B, Zhang L. The Role of Bone Marrow Microenvironment in Governing the Balance between Osteoblastogenesis and Adipogenesis. Aging Dis 2015; 7:514-25. [PMID: 27493836 DOI: 10.14336/ad.2015.1206] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 12/06/2015] [Indexed: 01/08/2023] Open
Abstract
In the adult bone marrow, osteoblasts and adipocytes share a common precursor called mesenchymal stem cells (MSCs). The plasticity between the two lineages has been confirmed over the past decades, and has important implications in the etiology of bone diseases such as osteoporosis, which involves an imbalance between osteoblasts and adipocytes. The commitment and differentiation of bone marrow (BM) MSCs is tightly controlled by the local environment that maintains a balance between osteoblast lineage and adipocyte. However, pathological conditions linked to osteoporosis can change the BM microenvironment and shift the MSC fate to favor adipocytes over osteoblasts, and consequently decrease bone mass with marrow fat accumulation. This review discusses the changes that occur in the BM microenvironment under pathological conditions, and how these changes affect MSC fate. We suggest that manipulating local environments could have therapeutic implications to avoid bone loss in diseases like osteoporosis.
Collapse
Affiliation(s)
- Jiao Li
- 1Department of Cell Biology, Zunyi Medical College, Zunyi, China
| | - Xingyu Liu
- 1Department of Cell Biology, Zunyi Medical College, Zunyi, China
| | - Bin Zuo
- 2Department of Orthopedic Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Li Zhang
- 3Department of Orthopedics, Tenth People's Hospital, Shanghai Tong Ji University, School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Li W, Wei S, Liu C, Song M, Wu H, Yang Y. Regulation of the osteogenic and adipogenic differentiation of bone marrow-derived stromal cells by extracellular uridine triphosphate: The role of P2Y2 receptor and ERK1/2 signaling. Int J Mol Med 2015; 37:63-73. [PMID: 26531757 PMCID: PMC4687443 DOI: 10.3892/ijmm.2015.2400] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/20/2015] [Indexed: 11/13/2022] Open
Abstract
An imbalance in the osteogenesis and adipogenesis of bone marrow-derived stromal cells (BMSCs) is a crucial pathological factor in the development of osteoporosis. Growing evidence suggests that extracellular nucleotide signaling involving the P2 receptors plays a significant role in bone metabolism. The aim of the present study was to investigate the effects of uridine triphosphate (UTP) on the osteogenic and adipogenic differentiation of BMSCs, and to elucidate the underlying mechanisms. The differentiation of the BMSCs was determined by measuring the mRNA and protein expression levels of osteogenic- and adipogenic-related markers, alkaline phosphatase (ALP) staining, alizarin red staining and Oil Red O staining. The effects of UTP on BMSC differentiation were assayed using selective P2Y receptor antagonists, small interfering RNA (siRNA) and an intracellular signaling inhibitor. The incubation of the BMSCs with UTP resulted in a dose-dependent decrease in osteogenesis and an increase in adipogenesis, without affecting cell proliferation. Significantly, siRNA targeting the P2Y2 receptor prevented the effects of UTP, whereas the P2Y6 receptor antagonist (MRS2578) and siRNA targeting the P2Y4 receptor had little effect. The activation of P2Y receptors by UTP transduced to the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway. This transduction was prevented by the mitogen-activated protein kinase inhibitor (U0126) and siRNA targeting the P2Y2 receptor. U0126 prevented the effects of UTP on osteogenic- and adipogenic-related gene expression after 24 h of culture, as opposed to 3 to 7 days of culture. Thus, our data suggest that UTP suppresses the osteogenic and enhances the adipogenic differentiation of BMSCs by activating the P2Y2 receptor. The ERK1/2 signaling pathway mediates the early stages of this process.
Collapse
Affiliation(s)
- Wenkai Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Sheng Wei
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Chaoxu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Mingyu Song
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| |
Collapse
|
34
|
Philbrick KA, Turner RT, Branscum AJ, Wong CP, Iwaniec UT. Paradoxical effects of partial leptin deficiency on bone in growing female mice. Anat Rec (Hoboken) 2015; 298:2018-29. [PMID: 26370912 DOI: 10.1002/ar.23267] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/25/2015] [Accepted: 03/24/2015] [Indexed: 11/09/2022]
Abstract
Morbidly obese, leptin-deficient ob/ob mice display low bone mass, mild osteoclast-rich osteopetrosis, and increased bone marrow adiposity. While partial leptin deficiency results in increased weight, the skeletal manifestations of partial leptin deficiency are less well defined. We therefore analyzed femora and lumbar vertebrae in growing (7-week-old) female C57BL/6 wildtype (WT) mice, partial leptin-deficient ob/+ mice, and leptin-deficient ob/ob mice. The bones were evaluated by dual energy absorptiometry, microcomputed tomography and histomorphometry. As expected, ob/+ mice were heavier, had more white adipose tissue, and lower serum leptin than WT mice, but were lighter and had less white adipose tissue than ob/ob mice. With a few exceptions, cancellous bone architecture, cell (osteoblast, osteoclast, and adipocyte), and dynamic measurements did not differ between WT and ob/+ mice. In contrast, compared to WT and ob/+ mice, ob/ob mice had lower cancellous bone volume fraction, and higher bone marrow adiposity in the femur metaphysis, and higher cancellous bone volume fraction in lumbar vertebra. Paradoxically, ob/+ mice had greater femoral bone volume than either WT or ob/ob mice. There was a positive correlation between body weight and femur volume in all three genotypes. However, the positive effect of weight on bone occurred with lower body weight in leptin-producing mice. The paradoxical differences in bone size among WT, ob/+, and ob/ob mice may be explained if leptin, in addition to stimulating bone growth and cancellous bone turnover, acts to lower the set-point at which increased body weight leads to a commensurate increase in bone size.
Collapse
Affiliation(s)
- Kenneth A Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, 97331, OR
| | - Russell T Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, 97331, OR.,Center for Healthy Aging Research, Oregon State University, Corvallis, 97331, OR
| | - Adam J Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, 97331, OR
| | - Carmen P Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, 97331, OR
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, 97331, OR.,Center for Healthy Aging Research, Oregon State University, Corvallis, 97331, OR
| |
Collapse
|
35
|
Chen XX, Yang T. Roles of leptin in bone metabolism and bone diseases. J Bone Miner Metab 2015; 33:474-85. [PMID: 25777984 DOI: 10.1007/s00774-014-0569-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 01/16/2014] [Indexed: 02/05/2023]
Abstract
Adipose tissue has been more accepted as an active contributor to whole body homeostasis, rather than just a fat depot, since leptin, a 16 kDa protein, was discovered as the product of the obese gene in 1994. With more and more studies conducted on this hormone, it has been shown that there is a close relationship between adipose tissue and bone, which have important effects on each other. Bone is the source of many hormones, such as osteocalcin, that can affect energy metabolism and then the anabolism or catabolism of fat tissue. In contrast, the adipose tissue synthesizes and releases a series of adipokines, which are involved in bone metabolism through direct or indirect effects on bone formation and resorption. Interestingly, leptin, one of the most important cytokines derived from fat tissue, seems to account for the largest part of effects on bone, through direct or indirect involvement in bone remodeling and by playing a significant role in many bone diseases, such as osteoporosis, osteoarthritis, rheumatic arthritis, bone tumors and even fractures. In this review, we will discuss the progress in leptin research, particularly focusing on the roles of leptin in bone diseases.
Collapse
Affiliation(s)
- Xu Xu Chen
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | | |
Collapse
|
36
|
Niu CC, Lin SS, Chen WJ, Liu SJ, Chen LH, Yang CY, Wang CJ, Yuan LJ, Chen PH, Cheng HY. Benefits of biphasic calcium phosphate hybrid scaffold-driven osteogenic differentiation of mesenchymal stem cells through upregulated leptin receptor expression. J Orthop Surg Res 2015; 10:111. [PMID: 26179165 PMCID: PMC4506435 DOI: 10.1186/s13018-015-0236-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/12/2015] [Indexed: 11/20/2022] Open
Abstract
Background The use of mesenchymal stem cells (MSCs) and coralline hydroxyapatite (HA) or biphasic calcium phosphate (BCP) as a bone substitute for posterolateral spinal fusion has been reported. However, the genes and molecular signals by which MSCs interact with their surrounding environment require further elucidation. Methods MSCs were harvested from bone grafting patients and identified by flow cytometry. A composite scaffold was developed using poly(lactide-co-glycolide) (PLGA) copolymer, coralline HA, BCP, and collagen as a carrier matrix for MSCs. The gene expression profiles of MSCs cultured in the scaffolds were measured by microarrays. The alkaline phosphatase (ALP) activity of the MSCs was assessed, and the expression of osteogenic genes and proteins was determined by quantitative polymerase chain reaction (Q-PCR) and Western blotting. Furthermore, we cultured rabbit MSCs in BCP or coralline HA hybrid scaffolds and transplanted these mixtures into rabbits for spinal fusion. We investigated the differences between BCP and coralline HA hybrid scaffolds by dual-energy X-ray absorptiometry (DEXA) and computed tomography (CT). Results Tested in vitro, the cells were negative for hematopoietic cell markers and positive for MSC markers. There was higher expression of 80 genes and lower of 101 genes of MSCs cultured in BCP hybrid scaffolds. Some of these genes have been shown to play a role in osteogenesis of MSCs. In addition, MSCs cultured in BCP hybrid scaffolds produced more messenger RNA (mRNA) for osteopontin, osteocalcin, Runx2, and leptin receptor (leptin-R) than those cultured in coralline HA hybrid scaffolds. Western blotting showed more Runx2 and leptin-R protein expression in BCP hybrid scaffolds. For in vivo results, 3D reconstructed CT images showed continuous bone bridges and fusion mass incorporated with the transverse processes. Bone mineral content (BMC) values were higher in the BCP hybrid scaffold group than in the coralline HA hybrid scaffold group. Conclusions The BCP hybrid scaffold for osteogenesis of MSCs is better than the coralline HA hybrid scaffold by upregulating expression of leptin-R. This was consistent with in vivo data, which indicated that BCP hybrid scaffolds induced more bone formation in a spinal fusion model.
Collapse
Affiliation(s)
- Chi-Chien Niu
- Department of Orthopaedics, Chang Gung Memorial Hospital, No. 5, Fu-Hsing Street 333, Kweishan, Taoyuan, Taiwan. .,College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Song-Shu Lin
- Department of Orthopaedics, Chang Gung Memorial Hospital, No. 5, Fu-Hsing Street 333, Kweishan, Taoyuan, Taiwan.
| | - Wen-Jer Chen
- Department of Orthopaedics, Chang Gung Memorial Hospital, No. 5, Fu-Hsing Street 333, Kweishan, Taoyuan, Taiwan. .,College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan, Taiwan.
| | - Lih-Huei Chen
- Department of Orthopaedics, Chang Gung Memorial Hospital, No. 5, Fu-Hsing Street 333, Kweishan, Taoyuan, Taiwan. .,College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Chuen-Yung Yang
- Department of Orthopaedics, Chang Gung Memorial Hospital, No. 5, Fu-Hsing Street 333, Kweishan, Taoyuan, Taiwan.
| | - Chao-Jan Wang
- Department of Radiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Li-Jen Yuan
- Department of Orthopaedics, Chang Gung Memorial Hospital, No. 5, Fu-Hsing Street 333, Kweishan, Taoyuan, Taiwan. .,College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Po-Han Chen
- Department of Orthopaedics, Chang Gung Memorial Hospital, No. 5, Fu-Hsing Street 333, Kweishan, Taoyuan, Taiwan.
| | - Hsiao-Yang Cheng
- Department of Radiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
37
|
Increased osteogenesis in osteoporotic bone marrow stromal cells by overexpression of leptin. Cell Tissue Res 2015; 361:845-56. [PMID: 25832621 DOI: 10.1007/s00441-015-2167-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 03/04/2015] [Indexed: 12/24/2022]
Abstract
Osteoporosis leads to increased bone fractures and net bone loss, in part because of the dysfunction of bone marrow stromal cells (BMSCs). Leptin is an adipokine that plays important roles in many biological processes, including the regulation of the actions of mesenchymal stem cells. Our aim is to investigate the osteogenic effects of leptin in osteoporotic BMSCs in vitro and in vivo. The leptin gene was transferred into BMSCs isolated from osteoporotic rats by using recombinant adenoviruses. Once the gene and protein expression of leptin had been confirmed, MTT assays were performed; leptin overexpression was confirmed not to affect the viability of osteoporotic BMSCs. However, alkaline phosphatase (ALP) activity measurements, Alizarin red staining and analyses by quantitative real-time reverse transcription with the polymerase chain reaction revealed that leptin upregulated ALP activity, mineral deposition and the mRNA levels of runt-related transcription factor 2, ALP and collagen type І. Lastly, the effects of leptin on osteogenic differentiation were assessed in vivo. Cells transfected with leptin exhibited increased osteogenic differentiation and enhanced formation of bone-like structures. This study thus reveals, for the first time, that the overexpression of leptin in osteoporotic BMSCs (1) enhances their capacity to differentiate into osteoblasts and to form bone-like tissue and (2) might be a useful skeletal regenerative therapy in osteoporotic patients.
Collapse
|
38
|
Granéli C, Karlsson C, Brisby H, Lindahl A, Thomsen P. The effects of PPAR-γ inhibition on gene expression and the progression of induced osteogenic differentiation of human mesenchymal stem cells. Connect Tissue Res 2014; 55:262-74. [PMID: 24708348 DOI: 10.3109/03008207.2014.910198] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mesenchymal stem cells (MSCs) can differentiate into several cell types, such as osteoblasts and adipocytes, both in vitro and in vivo. Although these two differentiation pathways are distinct from each other, cross-communication between cells of the two lineages exists both systemically and peripherally in the tissue. The transcription factor PPAR-γ, the main switch in adipogenic differentiation of MSCs, has previously been described to have a negative effect on osteogenic differentiation. The aim of this study was to investigate the effect of PPAR-γ inhibition on osteogenic differentiation of human MSCs, in vitro. Extracellular matrix analysis and quantification of osteogenic markers, revealed how these cells respond when the adipogenic differentiation pathway is blocked during induction of osteogenic differentiation. The inhibition leads to a significant increase in mineralization of the extracellular matrix, as well as an increased activity or up-regulated gene expression of alkaline phosphatase, the key enzyme involved in matrix mineralization. Furthermore, it was also demonstrated by microarray analysis, that PPAR-γ inhibition during osteogenic induction leads to a significant up-regulation of a number of genes related to both osteogenesis and adipogenesis such as c10orf10, leptin, GDF5 and KLF15. In conclusion, inhibition of PPAR-γ during induction of osteogenesis leads to increased osteogenic differentiation of human MSCs.
Collapse
Affiliation(s)
- Cecilia Granéli
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | | | | | | | | |
Collapse
|
39
|
Lindtner RA, Tiaden AN, Genelin K, Ebner HL, Manzl C, Klawitter M, Sitte I, von Rechenberg B, Blauth M, Richards PJ. Osteoanabolic effect of alendronate and zoledronate on bone marrow stromal cells (BMSCs) isolated from aged female osteoporotic patients and its implications for their mode of action in the treatment of age-related bone loss. Osteoporos Int 2014; 25:1151-61. [PMID: 23974861 DOI: 10.1007/s00198-013-2494-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 08/07/2013] [Indexed: 12/22/2022]
Abstract
SUMMARY In the present study, we evaluated the potential for aminobisphosphonates to enhance the development of bone-forming osteoblasts from progenitor cells isolated from aged female osteoporotic patients. The aminobisphosphonates tested significantly enhanced osteoblast formation and thus lend further insights into their possible mode of action in the treatment of osteoporosis. INTRODUCTION The primary aim of this study was to evaluate the influence of aminobisphosphonates on the osteogenesis of human bone marrow stromal cells (hBMSCs) and mineralization of differentiating bone-forming cells isolated from osteoporotic patients. METHODS The influence of aminobisphosphonate treatment on hBMSC osteogenesis was assessed by the quantitative measurement of alkaline phosphatase (ALP) activity, in addition to quantitative reverse transcription polymerase chain reaction and Western blot analysis of known osteogenic markers. Mineralized matrix formation by hBMSC-derived osteoblasts was visualized and quantified using Alizarin red staining. RESULTS hBMSC cultures treated with osteogenic medium supplemented with zoledronate demonstrated a significant increase in Alizarin red staining after 3 weeks as compared to cells cultured in osteogenic medium alone. Similarly, cultures of differentiating hBMSCs isolated from patients receiving alendronate treatment also demonstrated an increased propensity for mineralization, even in the absence of further in vitro stimulation by zoledronate. The stimulatory effects of aminobisphosphonate treatment on hBMSC-derived osteoblast-mediated mineralization were independent of any alterations in ALP activity, although significant decreases in the expression levels of osteopontin (SPP1) were evident in hBMSCs following exposure to aminobisphosphonates. Further analysis including Western blotting and loss-of-function studies revealed osteopontin as having a negative influence on the mineralization of differentiating osteoporotic bone-forming cells. CONCLUSIONS The results presented here demonstrate for the first time that aminobisphosphonate treatment of osteoporotic hBMSCs enhances their capacity for osteoblast formation and subsequent mineral deposition, thus supporting the concept of aminobisphosphonates as having an osteoanabolic effect in osteoporosis.
Collapse
Affiliation(s)
- R A Lindtner
- Department of Trauma Surgery and Sports Medicine, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Are volumetric bone mineral density and bone micro-architecture associated with leptin and soluble leptin receptor levels in adolescent idiopathic scoliosis?--A case-control study. PLoS One 2014; 9:e87939. [PMID: 24516571 PMCID: PMC3916359 DOI: 10.1371/journal.pone.0087939] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 01/03/2014] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Adolescent idiopathic scoliosis (AIS) is associated with low bone mineral density (BMD). The underlying etiology and how it may relate to the development of osteopenia remains unknown. Leptin has been postulated as one of the etiologic factors of AIS because of its profound effects on bone metabolism and pubertal growth. Its modulator, soluble leptin receptor (sOB-R), may affect leptin bioavailability and signaling. This study aimed to investigate whether serum leptin and sOB-R levels may be associated with bone quality, and whether these relationships may differ between young adolescent girls with and without AIS. METHODS This was a case-control study involving 94 newly diagnosed AIS girls (Cobb angle 12-48°) aged 12 to 14 years old and 87 age and gender-matched normal controls. Subjects with BMI>23.0 Kg/m(2) were excluded. Anthropometric measurements including body weight, height, arm span and sitting height were taken. Serum total leptin and sOB-R were assayed with ELISA. Non-dominant distal radius was scanned with High Resolution pQCT for assessing bone quality in terms of bone morphometry, volumetric BMD (vBMD) and trabecular bone micro-architecture. RESULTS Compared with normal controls, AIS girls had numerically higher sOB-R (p = 0.006), lower average vBMD (p = 0.048), lower cortical vBMD (p = 0.029), higher cortical bone perimeter (p = 0.014) and higher trabecular area (p = 0.027), but none remained statistically significant after the Hochberg-Benjamini procedure. Correlation analysis on serum leptin level indicated that distinctive correlations with trabecular bone parameters occurred only in AIS. CONCLUSION This study showed that bone quality in AIS girls was deranged as compared with controls. In addition, the distinct differences in correlation pattern between leptin and trabecular bone parameters indicated possible abnormalities in bone metabolism and dysfunction of the leptin signaling pathway in AIS.
Collapse
|
41
|
Boyette LB, Creasey OA, Guzik L, Lozito T, Tuan RS. Human bone marrow-derived mesenchymal stem cells display enhanced clonogenicity but impaired differentiation with hypoxic preconditioning. Stem Cells Transl Med 2014; 3:241-54. [PMID: 24436440 DOI: 10.5966/sctm.2013-0079] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells are promising candidate cells for regenerative applications because they possess high proliferative capacity and the potential to differentiate into other cell types. Mesenchymal stem cells (MSCs) are easily sourced but do not retain their proliferative and multilineage differentiative capabilities after prolonged ex vivo propagation. We investigated the use of hypoxia as a preconditioning agent and in differentiating cultures to enhance MSC function. Culture in 5% ambient O(2) consistently enhanced clonogenic potential of primary MSCs from all donors tested. We determined that enhanced clonogenicity was attributable to increased proliferation, increased vascular endothelial growth factor secretion, and increased matrix turnover. Hypoxia did not impact the incidence of cell death. Application of hypoxia to osteogenic cultures resulted in enhanced total mineral deposition, although this effect was detected only in MSCs preconditioned in normoxic conditions. Osteogenesis-associated genes were upregulated in hypoxia, and alkaline phosphatase activity was enhanced. Adipogenic differentiation was inhibited by exposure to hypoxia during differentiation. Chondrogenesis in three-dimensional pellet cultures was inhibited by preconditioning with hypoxia. However, in cultures expanded under normoxia, hypoxia applied during subsequent pellet culture enhanced chondrogenesis. Whereas hypoxic preconditioning appears to be an excellent way to expand a highly clonogenic progenitor pool, our findings suggest that it may blunt the differentiation potential of MSCs, compromising their utility for regenerative tissue engineering. Exposure to hypoxia during differentiation (post-normoxic expansion), however, appears to result in a greater quantity of functional osteoblasts and chondrocytes and ultimately a larger quantity of high-quality differentiated tissue.
Collapse
Affiliation(s)
- Lisa B Boyette
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, McGowan Institute for Regenerative Medicine, and Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
42
|
|
43
|
|
44
|
Turner RT, Kalra SP, Wong CP, Philbrick KA, Lindenmaier LB, Boghossian S, Iwaniec UT. Peripheral leptin regulates bone formation. J Bone Miner Res 2013; 28:22-34. [PMID: 22887758 PMCID: PMC3527690 DOI: 10.1002/jbmr.1734] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 06/30/2012] [Accepted: 07/16/2012] [Indexed: 12/15/2022]
Abstract
Substantial evidence does not support the prevailing view that leptin, acting through a hypothalamic relay, decreases bone accrual by inhibiting bone formation. To clarify the mechanisms underlying regulation of bone architecture by leptin, we evaluated bone growth and turnover in wild-type (WT) mice, leptin receptor-deficient db/db mice, leptin-deficient ob/ob mice, and ob/ob mice treated with leptin. We also performed hypothalamic leptin gene therapy to determine the effect of elevated hypothalamic leptin levels on osteoblasts. Finally, to determine the effects of loss of peripheral leptin signaling on bone formation and energy metabolism, we used bone marrow (BM) from WT or db/db donor mice to reconstitute the hematopoietic and mesenchymal stem cell compartments in lethally irradiated WT recipient mice. Decreases in bone growth, osteoblast-lined bone perimeter and bone formation rate were observed in ob/ob mice and greatly increased in ob/ob mice following subcutaneous administration of leptin. Similarly, hypothalamic leptin gene therapy increased osteoblast-lined bone perimeter in ob/ob mice. In spite of normal osteoclast-lined bone perimeter, db/db mice exhibited a mild but generalized osteopetrotic-like (calcified cartilage encased by bone) skeletal phenotype and greatly reduced serum markers of bone turnover. Tracking studies and histology revealed quantitative replacement of BM cells following BM transplantation. WT mice engrafted with db/db BM did not differ in energy homeostasis from untreated WT mice or WT mice engrafted with WT BM. Bone formation in WT mice engrafted with WT BM did not differ from WT mice, whereas bone formation in WT mice engrafted with db/db cells did not differ from the low rates observed in untreated db/db mice. In summary, our results indicate that leptin, acting primarily through peripheral pathways, increases osteoblast number and activity.
Collapse
Affiliation(s)
- Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Satya P. Kalra
- Department of Neuroscience, University of Florida McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Carmen P. Wong
- Molecular and Cellular Nutrition Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Kenneth A. Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Laurence B. Lindenmaier
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Stephane Boghossian
- Department of Neuroscience, University of Florida McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
45
|
Jin WJ, Jiang SD, Jiang LS, Dai LY. Differential responsiveness to 17β-estradiol of mesenchymal stem cells from postmenopausal women between osteoporosis and osteoarthritis. Osteoporos Int 2012; 23:2469-78. [PMID: 22159632 DOI: 10.1007/s00198-011-1859-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 11/22/2011] [Indexed: 02/05/2023]
Abstract
UNLABELLED Differential osteogenic potential and responsiveness to 17β-estradiol (E2) of mesenchymal stem cells (MSCs) were found between postmenopausal women with osteoporosis (OP) and osteoarthritis (OA). These results suggest differential biological mechanisms of estrogen deficiency in regulation of bone remodeling between OP and OA. INTRODUCTION OP and OA are two common disorders in postmenopausal women. The inverse relationship has been suggested between OP and OA, but their mechanisms that relate to estrogen deficiency are not fully understood. The aim of this study was to compare the differential responsiveness to E2 of MSCs from osteoporotic versus osteoarthritic donors. METHODS Twenty postmenopausal patients, ten with osteoporotic hip fractures and ten with hip osteoarthritis, were included into this study. MSCs were derived from cancellous bones of femoral heads from OA and OP donors and cultured in osteogenic and adipogenic medium with or without E2 added. The alkaline phosphatase (ALP) activity, calcium content, calcified nodules, lipid droplets, messenger RNA (mRNA) expression of ALP, osteocalcin (OC), collagen 1α (COL1α), peroxisome proliferators-activated receptor γ2 (PPARγ2) and lipoprotein lipase (LPL) were measured and compared between two groups with OP and OA. RESULTS In osteogenic medium, ALP activity, calcium content and mRNA expression of OC and COL1α in MSCs from OA were significantly higher than those from OP group. In adipogenic condition, there was no significant difference in lipid droplets formation and mRNA expression of PPARγ2 and LPL between OP and OA groups. With E2 added in osteogenic medium, ALP activity, calcium content and OC mRNA were significantly higher in OP group than in OA group, whereas E2 had no significant effect on lipid droplet formation and mRNA expression of PPARγ2 and LPL. CONCLUSION Differential osteogenic potential and responsiveness to E2 of MSCs were found between postmenopausal women with OP and OA. These results may provide information for clinical application of MSCs in the differential setting of estrogen deficiency.
Collapse
Affiliation(s)
- W-J Jin
- Department of Orthopaedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
46
|
Abstract
Osteoporosis and obesity are chronic disorders that are both increasing in prevalence. The pathophysiology of these conditions is multifactorial and includes genetic, environmental and hormonal determinants. Although it has long been considered that these are distinct disorders rarely found in the same individual, emerging evidence from basic and clinical studies support an important interaction between adipose tissue and the skeleton. It is proposed that adiposity may influence bone remodelling through three mechanisms: (i) secretion of cytokines that directly target bone, (ii) production of adipokines that influence the central nervous system thereby changing sympathetic impulses to bone and (iii) paracrine influences on adjacent skeletal cells. Here we focus on the current understanding of bone-fat interactions and the clinical implications of recent studies linking obesity to osteoporosis.
Collapse
Affiliation(s)
- M Kawai
- Department of Bone and Mineral Research, Osaka Medical Center and Research Institute for Maternal and Child Health, Izumi, Osaka, Japan
| | | | | |
Collapse
|
47
|
Churchman SM, Ponchel F, Boxall SA, Cuthbert R, Kouroupis D, Roshdy T, Giannoudis PV, Emery P, McGonagle D, Jones EA. Transcriptional profile of native CD271+ multipotential stromal cells: evidence for multiple fates, with prominent osteogenic and Wnt pathway signaling activity. ARTHRITIS AND RHEUMATISM 2012; 64:2632-43. [PMID: 22378497 DOI: 10.1002/art.34434] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Controversy surrounds the identity and functionality of rare bone marrow-derived multipotential stromal cells (BM-MSCs), including their differentiation capabilities, their relationship to pericytes and hematopoiesis-supporting stromal cells, and the relevance of their culture-expanded progeny in studies of skeletal biology and development of cell-based therapies. The aim of this study was to clarify the nature of candidate BM-MSCs by profiling transcripts that reflect different aspects of their putative functions in vivo. METHODS Rare, sorted BM-derived CD45(-/low) CD271(bright) (CD271) cells were analyzed using 96-gene expression arrays focused on transcripts relevant to mesenchymal-lineage differentiation (toward bone, cartilage, fat, or muscle), hematopoietic and stromal support, and molecules critical to skeletal homeostasis. These cells were compared to matched CD45+ CD271- hematopoietic-lineage cells, culture-expanded MSCs, and skin fibroblasts. When feasible, transcription was validated using flow cytometry. RESULTS CD271 cells had a transcriptional profile consistent with the multiple fates of in vivo MSCs, evident from the observed simultaneous expression of osteogenic, adipogenic, pericytic, and hematopoiesis-supporting genes (e.g., SP7 [osterix], FABP4 [fatty acid binding protein 4], ANGPT1 [angiopoietin 1], and CXCL12 [stromal cell-derived factor 1], respectively). Compared to culture-expanded MSCs and fibroblasts, CD271 cells exhibited greater transcriptional activity, particularly with respect to Wnt-related genes (>1,000-fold increased expression of FRZB [secreted frizzled-related protein 3] and WIF1 [Wnt inhibitory factor 1]). A number of transcripts were identified as novel markers of MSCs. CONCLUSION The native, BM-derived in vivo MSC population is endowed with a gene signature that is compatible with multiple functions, reflecting the topographic bone niche of these cells, and their signature is significantly different from that of culture-expanded MSCs. This indicates that studies of the biologic functions of MSCs in musculoskeletal diseases, including osteoporosis and osteoarthritis, should focus on in vivo MSCs, rather than their culture-adapted progeny.
Collapse
Affiliation(s)
- Sarah M Churchman
- NIHR Leeds Musculoskeletal Biomedical Research Unit and University of Leeds, Leeds, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Pino AM, Rosen CJ, Rodríguez JP. In osteoporosis, differentiation of mesenchymal stem cells (MSCs) improves bone marrow adipogenesis. Biol Res 2012; 45:279-87. [PMID: 23283437 PMCID: PMC8262098 DOI: 10.4067/s0716-97602012000300009] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 08/23/2012] [Indexed: 01/01/2023] Open
Abstract
The formation, maintenance, and repair of bone tissue involve close interlinks between two stem cell types housed in the bone marrow: the hematologic stem cell originating osteoclasts and mesenchymal stromal cells (MSCs) generating osteoblasts. In this review, we consider malfunctioning of MSCs as essential for osteoporosis. In osteoporosis, increased bone fragility and susceptibility to fractures result from increased osteoclastogenesis and insufficient osteoblastogenesis. MSCs are the common precursors for both osteoblasts and adipocytes, among other cell types. MSCs' commitment towards either the osteoblast or adipocyte lineages depends on suitable regulatory factors activating lineage-specific transcriptional regulators. In osteoporosis, the reciprocal balance between the two differentiation pathways is altered, facilitating adipose accretion in bone marrow at the expense of osteoblast formation; suggesting that under this condition MSCs activity and their microenvironment may be disturbed. We summarize research on the properties of MSCs isolated from the bone marrow of control and osteoporotic post-menopausal women. Our observations indicate that intrinsic properties of MSCs are disturbed in osteoporosis. Moreover, we found that the regulatory conditions in the bone marrow fluid of control and osteoporotic patients are significantly different. These conclusions should be relevant for the use of MSCs in therapeutic applications.
Collapse
Affiliation(s)
- Ana María Pino
- Laboratorio de Biología Celular y Molecular, INTA, Universidad de Chile
| | | | | |
Collapse
|
49
|
Interleukin-17A increases leptin production in human bone marrow mesenchymal stem cells. Biochem Pharmacol 2011; 83:661-70. [PMID: 22197587 DOI: 10.1016/j.bcp.2011.12.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/08/2011] [Accepted: 12/09/2011] [Indexed: 02/07/2023]
Abstract
Lineage commitment of human bone marrow mesenchymal stem cells (hBM-MSCs) to adipocytes or osteoblasts has been suggested as a model system to study the relationship between type II diabetes and abnormal bone metabolism. Leptin and IL-17A inhibit adipogenesis whereas they promote osteogenesis in MSCs. Due to pathophysiologic roles of IL-17A in human metabolic diseases and bone metabolism, it was evaluated whether IL-17A-dependent inverse regulation on adipogenesis and osteogenesis was related to endogenous leptin production in hBM-MSCs. In the analysis of adiponectin and leptin secretion profiles of hBM-MSCs in response to various combinations of differentiation inducing factors, it was found that dexamethasone, a common molecule used for both adipogenesis and osteogenesis, increased leptin production in hBM-MSCs. Importantly, the level of leptin production during osteogenesis in hBM-MSCs was higher than that during adipogenesis, implicating a significant leptin production in extra-adipose tissues. IL-17A increased leptin production in hBM-MSCs and also under the condition of osteogenesis. In spite of direct inhibition on adipogenesis, IL-17A up-regulated leptin production in hBM-MSC-derived adipocytes. Anti-leptin antibody treatment partially antagonized the IL-17A dependent inhibition of adipogenesis in hBM-MSCs, suggesting a role of leptin in mediating the inverse regulation of IL-17A on osteogenesis and adipogenesis in hBM-MSCs. Therefore, the IL-17A-induced leptin production may provide a key clue to understand a molecular mechanism on the lineage commitment of hBM-MSCs into adipocytes or osteoblasts. In addition, leptin production in extra-adipose tissues like MSCs and osteoblasts should be considered in future studies on leptin-associated human diseases.
Collapse
|
50
|
Scheller EL, Song J, Dishowitz MI, Soki FN, Hankenson KD, Krebsbach PH. Leptin functions peripherally to regulate differentiation of mesenchymal progenitor cells. Stem Cells 2010; 28:1071-80. [PMID: 20506495 DOI: 10.1002/stem.432] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Leptin functions through a well-documented central neuroendocrine pathway to regulate bone mass. However, the ability of leptin to modulate bone mass through a peripheral mechanism has been debated due to conflicting in vitro results and lack of sufficient in vivo models. We utilized mice with LoxP sites introduced into the long-form leptin receptor (ObRb) gene to determine how leptin regulates mesenchymal progenitor cell (MPC) differentiation and osteoblast function in vitro and in vivo. Rapid phosphorylation of Stat3 after leptin treatment of bone marrow stromal cells (BMSCs) from mice with conditional deletion of ObRb in macrophages (LysM(Cre+F/F)) confirmed expression of functional leptin receptors by BMSCs. Adenovirus-Cre mediated disruption of ObRb in primary stromal cells decreased mineralization and increased adipogenesis. In contrast, BMSCs harvested from leptin-signaling deficient Ob/Ob or Db/Db mice showed increased mineralization. To determine the physiologic relevance of these differences, mice with cell-specific deletion of ObRb in mesenchymal precursors (3.6(Cre+F/F)) or osteoblasts (2.3(Cre+F/F)) were generated. Although the 2.3(Cre+F/F) mice were grossly normal, the 3.6(Cre+F/F) mice displayed mild obesity that was not attributed to food intake. Femurs of 3.6(Cre+F/F) animals showed a 58%-61.9% increase in trabecular bone volume and a 65.5%-74% increase in bone mineral density. Cortical volume and mineral content were also increased 18%-22%. Primary 3.6(Cre+F/F) BMSCs recapitulated the high mineralization phenotype of Ob/Ob and Db/Db BMSCs. We conclude that leptin may have multiple peripheral roles depending on the differentiation state of MPC. Leptin (a) helps maintain MPCs in an undifferentiated state and (b) promotes mineralization of more differentiated osteoblasts.
Collapse
Affiliation(s)
- Erica L Scheller
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | | | | | | | | | | |
Collapse
|