1
|
Liu H, Liu L, Rosen CJ. Bone Marrow Adipocytes as Novel Regulators of Metabolic Homeostasis: Clinical Consequences of Bone Marrow Adiposity. Curr Obes Rep 2025; 14:9. [PMID: 39808256 DOI: 10.1007/s13679-024-00594-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW Bone marrow adipose tissue is a distinctive fat depot located within the skeleton, with the potential to influence both local and systemic metabolic processes. Although significant strides have been made in understanding bone marrow adipose tissue over the past decade, many questions remain regarding their precise lineage and functional roles. RECENT FINDINGS Recent studies have highlighted bone marrow adipose tissue's involvement in continuous cross-talk with other organs and systems, exerting both endocrine and paracrine functions that play a crucial role in metabolic homeostasis, skeletal remodeling, hematopoiesis, and the progression of bone metastases. The advancement of imaging techniques, particularly cross-sectional imaging, has profoundly expanded our understanding of the complexities beyond the traditional view of bone marrow adipose tissue as an inert depot. Notably, marrow adipocytes are anatomically and functionally distinct from brown, beige, and classic white adipocytes. Emerging evidence suggests that bone marrow adipocytes, bone marrow adipose tissue originate from the differentiation of bone marrow mesenchymal stromal cells; however, they appear to be a heterogeneous population with varying metabolic profiles, lipid compositions, secretory properties, and functional responses depending on their specific location within the bone marrow. This review provides an up-to-date synthesis of current knowledge on bone marrow adipocytes, emphasizing the relationships between bone marrow adipogenesis and factors such as aging, osteoporosis, obesity, and bone marrow tumors or metastases, thereby elucidating the mechanisms underlying musculoskeletal pathophysiology.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology &, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Linyi Liu
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA.
| |
Collapse
|
2
|
Schwartzman JD, McCall M, Ghattas Y, Pugazhendhi AS, Wei F, Ngo C, Ruiz J, Seal S, Coathup MJ. Multifunctional scaffolds for bone repair following age-related biological decline: Promising prospects for smart biomaterial-driven technologies. Biomaterials 2024; 311:122683. [PMID: 38954959 DOI: 10.1016/j.biomaterials.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
The repair of large bone defects due to trauma, disease, and infection can be exceptionally challenging in the elderly. Despite best clinical practice, bone regeneration within contemporary, surgically implanted synthetic scaffolds is often problematic, inconsistent, and insufficient where additional osteobiological support is required to restore bone. Emergent smart multifunctional biomaterials may drive important and dynamic cellular crosstalk that directly targets, signals, stimulates, and promotes an innate bone repair response following age-related biological decline and when in the presence of disease or infection. However, their role remains largely undetermined. By highlighting their mechanism/s and mode/s of action, this review spotlights smart technologies that favorably align in their conceivable ability to directly target and enhance bone repair and thus are highly promising for future discovery for use in the elderly. The four degrees of interactive scaffold smartness are presented, with a focus on bioactive, bioresponsive, and the yet-to-be-developed autonomous scaffold activity. Further, cell- and biomolecular-assisted approaches were excluded, allowing for contemporary examination of the capabilities, demands, vision, and future requisites of next-generation biomaterial-induced technologies only. Data strongly supports that smart scaffolds hold significant promise in the promotion of bone repair in patients with a reduced osteobiological response. Importantly, many techniques have yet to be tested in preclinical models of aging. Thus, greater clarity on their proficiency to counteract the many unresolved challenges within the scope of aging bone is highly warranted and is arguably the next frontier in the field. This review demonstrates that the use of multifunctional smart synthetic scaffolds with an engineered strategy to circumvent the biological insufficiencies associated with aging bone is a viable route for achieving next-generation therapeutic success in the elderly population.
Collapse
Affiliation(s)
| | - Max McCall
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Yasmine Ghattas
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Fei Wei
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA; Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, USA, Orlando, FL
| | - Melanie J Coathup
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
3
|
Xu J, Huang Z, Shi S, Xia J, Chen G, Zhou K, Zhang Y, Bian C, Shen Y, Yin X, Lu L, Gu H. Glial maturation factor-β deficiency prevents oestrogen deficiency-induced bone loss by remodelling the actin network to suppress adipogenesis of bone marrow mesenchymal stem cells. Cell Death Dis 2024; 15:829. [PMID: 39543090 PMCID: PMC11564563 DOI: 10.1038/s41419-024-07234-z] [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: 07/27/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
An imbalance between the adipogenesis and osteogenesis of bone marrow mesenchymal stem cells (BMSCs) is considered the basic pathogenesis of osteoporosis. Although actin cytoskeleton remodelling plays a crucial role in the differentiation of BMSCs, the role of actin cytoskeleton remodelling in the adipogenesis of BMSCs and postmenopausal osteoporosis (PMOP) has remained elusive. Glia maturation factor-beta (GMFB) has a unique role in remodelling the polymerization/depolymerization cycles of actin. We observed that GMFB expression was increased in bone tissue from both ovariectomized (OVX) rats and PMOP patients. GMFB knockout inhibited the accumulation of bone marrow adipocytes and increased bone mass in the OVX rat model. The inhibition of adipocyte differentiation in GMFB knockout BMSCs was mediated via actin cytoskeleton remodelling and the Ca2+-calcineurin-NFATc2 axis. Furthermore, we found that GMFB shRNA treatment in vivo had favourable effects on osteoporosis induced by OVX. Together, these findings suggest a pathological association of the GMFB with PMOP and highlight the potential of the GMFB as a therapeutic target for osteoporosis patients.
Collapse
Affiliation(s)
- Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Si Shi
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China
| | - Jiangni Xia
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Guangnan Chen
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Yiming Zhang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Chong Bian
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Yuqin Shen
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China.
| | - Lixia Lu
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China.
- Department of Biochemistry and Molecular Biology, Tongji University School of medicine, Shanghai, PR China.
| | - Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
4
|
Niu H, Zhou M, Xu X, Xu X. Bone Marrow Adipose Tissue as a Critical Regulator of Postmenopausal Osteoporosis - A Concise Review. Clin Interv Aging 2024; 19:1259-1272. [PMID: 39011312 PMCID: PMC11249116 DOI: 10.2147/cia.s466446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a major health problem affecting millions of women worldwide. PMOP patients are often accompanied by abnormal accumulation of bone marrow adipose tissue (BMAT). BMAT is a critical regulator of bone homeostasis, and an increasing BMAT volume is negatively associated with bone mass reduction or fracture. BMAT regulates bone metabolism via adipokines, cytokines and the immune system, but the specific mechanisms are largely unknown. This review emphasizes the impact of estrogen deficiency on bone homeostasis and BMAT expansion, and the mechanism by which BMAT regulates PMOP, providing a promising strategy for targeting BMAT in preventing and treating PMOP.
Collapse
Affiliation(s)
- Huifang Niu
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Minfeng Zhou
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaoyun Xu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, People's Republic of China
| | - Xiaojuan Xu
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
5
|
Yang Z, Liu C, Shi Z, Qin J. IDEAL-IQ combined with intravoxel incoherent motion diffusion-weighted imaging for quantitative diagnosis of osteoporosis. BMC Med Imaging 2024; 24:155. [PMID: 38902641 PMCID: PMC11188172 DOI: 10.1186/s12880-024-01326-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Osteoporosis (OP) is a common chronic metabolic bone disease characterized by decreased bone mineral content and microstructural damage, leading to increased fracture risk. Traditional methods for measuring bone density have limitations in accurately distinguishing vertebral bodies and are influenced by vertebral degeneration and surrounding tissues. Therefore, novel methods are needed to quantitatively assess changes in bone density and improve the accurate diagnosis of OP. METHODS This study aimed to explore the applicative value of the iterative decomposition of water and fat with echo asymmetry and least-squares estimation-iron (IDEAL-IQ) sequence combined with intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) for the diagnosis of osteoporosis. Data from 135 patients undergoing dual-energy X-ray absorptiometry (DXA), IDEAL-IQ, and IVIM-DWI were prospectively collected and analyzed. Various parameters obtained from IVIM-DWI and IDEAL-IQ sequences were compared, and their diagnostic efficacy was evaluated. RESULTS Statistically significant differences were observed among the three groups for FF, R2*, f, D, DDC values, and BMD values. FF and f values exhibited negative correlations with BMD values, with r=-0.313 and - 0.274, respectively, while R2*, D, and DDC values showed positive correlations with BMD values, with r = 0.327, 0.532, and 0.390, respectively. Among these parameters, D demonstrated the highest diagnostic efficacy for osteoporosis (AUC = 0.826), followed by FF (AUC = 0.713). D* exhibited the lowest diagnostic performance for distinguishing the osteoporosis group from the other two groups. Only D showed a significant difference between genders. The AUCs for IDEAL-IQ, IVIM-DWI, and their combination were 0.74, 0.89, and 0.90, respectively. CONCLUSIONS IDEAL-IQ combined with IVIM-DWI provides valuable information for the diagnosis of osteoporosis and offers evidence for clinical decisions. The superior diagnostic performance of IVIM-DWI, particularly the D value, suggests its potential as a more sensitive and accurate method for diagnosing osteoporosis compared to IDEAL-IQ. These findings underscore the importance of integrating advanced imaging techniques into clinical practice for improved osteoporosis management and highlight the need for further research to explore the full clinical implications of these imaging modalities.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Chenglong Liu
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Zhaojuan Shi
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Jian Qin
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China.
| |
Collapse
|
6
|
Lu PY, Huang M, Shao MH, Hu JX, Ding CY, Feng YJ, Zhang M, Lin HP, Tian HS. Effect and mechanism of recombinant human fibroblast growth factor 18 on osteoporosis in OVX mice. Climacteric 2024; 27:305-313. [PMID: 38275172 DOI: 10.1080/13697137.2024.2302967] [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: 09/27/2022] [Accepted: 12/29/2023] [Indexed: 01/27/2024]
Abstract
OBJECTIVES This study aimed to investigate the effect and the mechanism of recombinant human fibroblast growth factor 18 (rhFGF18) on postmenopausal osteoporosis. METHODS The effect of rhFGF18 on the proliferation and apoptosis of osteoblasts and the mechanism underlying such an effect was evaluated using an oxidative stress model of the MC3T3-E1 cell line. Furthermore, ovariectomy was performed on ICR mice to imitate estrogen-deficiency postmenopausal osteoporosis. Bone metabolism and bone morphological parameters in the ovariectomized (OVX) mice were evaluated. RESULTS The results obtained from the cell model showed that FGF18 promoted MC3T3-E1 cell proliferation by activating the extracellular signal-regulated kinase (ERK) and p38 instead of c-Jun N-terminal kinase (JNK). FGF18 also prevented cells from damage inflicted by oxidative stress via inhibition of apoptosis. After FGF18 administration, the expression level of anti-apoptotic protein Bcl-2 in the mice was upregulated, whereas those of the pro-apoptotic proteins Bax and caspase-3 were downregulated. Administering FGF18 also improved bone metabolism and bone morphological parameters in OVX mice. CONCLUSIONS FGF18 could effectively prevent bone loss in OVX mice by enhancing osteoblastogenesis and protecting osteoblasts from oxidative stress-induced apoptosis.
Collapse
Affiliation(s)
- P Y Lu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - M Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
- Department of Pharmacy, Wuzhou GongRen Hospital, Wuzhou, China
| | - M H Shao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - J X Hu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - C Y Ding
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Y J Feng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - M Zhang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - H P Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - H S Tian
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Dong H, Tang F, Zhao Z, Huang W, Wan X, Hong Z, Liu Y, Dong X, Chen S. The Bioactive Compounds of Epimedium and Their Potential Mechanism of Action in Treating Osteoporosis: A Network Pharmacology and Experimental Validation Study. Pharmaceuticals (Basel) 2024; 17:706. [PMID: 38931373 PMCID: PMC11206986 DOI: 10.3390/ph17060706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Osteoporosis is a global health challenge characterized by bone loss and microstructure deterioration, which urgently requires the development of safer and more effective treatments due to the significant adverse effects and limitations of existing drugs for long-term treatment. Traditional Chinese medicine, like Epimedium, offers fewer side effects and has been used to treat osteoporosis, yet its active compounds and pharmacological mechanisms remain unclear. In this study, 65 potential active compounds, 258 potential target proteins, and 488 pathways of Epimedium were identified through network pharmacology analysis. Further network analysis and review of the literature identified six potential active compounds and HIF-1α for subsequent experimental validation. In vitro experiments confirmed that 2″-O-RhamnosylIcariside II is the most effective compound among the six potential active compounds. It can promote osteoblast differentiation, bind with HIF-1α, and inhibit both HIF-1α gene and protein expression, as well as enhance COL1A1 protein expression under hypoxic conditions. In vivo experiments demonstrated its ability to improve bone microstructures and reduce bone loss by decreasing bone marrow adipose tissue, enhancing bone formation, and suppressing HIF-1α protein expression. This study is the first to describe the therapeutic effects of 2-O-RhamnosylIcariside II on osteoporosis, which was done, specifically, through a mechanism that targets and inhibits HIF-1α. This study provides a scientific basis for the clinical application of Epimedium and offers a new candidate drug for the treatment of osteoporosis. Additionally, it provides new evidence supporting HIF-1α as a therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Huizhong Dong
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Fen Tang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Zilu Zhao
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Wenxuan Huang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xiangyang Wan
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Zhanying Hong
- School of Pharmacy, Naval Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Ying Liu
- Institute of Translational Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China;
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Si Chen
- School of Medicine, Shanghai University, Shanghai 200444, China
| |
Collapse
|
8
|
Li J, Lu L, Liu L, Wang C, Xie Y, Li H, Tian L, Yu X. The unique role of bone marrow adipose tissue in ovariectomy-induced bone loss in mice. Endocrine 2024; 83:77-91. [PMID: 37682419 DOI: 10.1007/s12020-023-03504-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Accumulation of bone marrow adipose tissue (BMAT) is always seen in osteoporosis induced by estrogen deficiency. Herein, we aimed to investigate the mechanisms and consequences of this phenomenon by establishing a mouse model of osteoporosis caused by ovariectomy (OVX)-mimicked estrogen deficiency. METHODS Micro-CT, osmium tetroxide staining, and histological analyses were performed to examine the changes in bone microstructure, BMAT and white adipose tissue (WAT) in OVX mice compared to sham mice. The osteogenesis and adipogenesis of primary bone marrow stromal cells (BMSCs) isolated from sham and OVX mice were compared in vitro. The molecular phenotypes of BMAT and WAT were determined and compared by quantitative PCR (qPCR). Bone marrow adipocyte-conditioned medium (BMA CM) was prepared from sham or OVX mice for coculture assays, and BMSCs or bone marrow monocytes/macrophages (BMMs) were isolated and subjected to osteoblast and osteoclast differentiation, respectively. Cell staining and qPCR were used to assess the effects of BMAT on bone metabolism. RESULTS OVX-induced estrogen deficiency induced reductions in both cortical and trabecular bone mass along with an expansion of BMAT volume. At the cellular level, loss of estrogen inhibited BMSC osteogenesis and promoted BMSC adipogenesis, whereas addition of estradiol exerted the opposite effects. In response to estrogen deficiency, despite the common proinflammatory molecular phenotype observed in both fat depots, BMAT, unlike WAT, unexpectedly exhibited an increase in adipocyte differentiation and lipolytic activity as well as the maintenance of insulin sensitivity. Importantly, BMAT, but not WAT, presented increased mRNA levels of both BMP receptor inhibitors (Grem1, Chrdl1) and Rankl following OVX. In addition, treatment with BMA CM, especially from OVX mice, suppressed the osteoblast differentiation of BMSCs while favoring the osteoclast differentiation of BMMs. CONCLUSION Our study illustrates that OVX-induced estrogen deficiency results in bone loss and BMAT expansion by triggering imbalance between the osteogenesis and adipogenesis of BMSCs. Furthermore, expanded BMAT, unlike typical WAT, may negatively regulate bone homeostasis through paracrine inhibition of osteoblast-mediated bone formation and promotion of osteoclast-mediated bone resorption.
Collapse
Affiliation(s)
- Jiao Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lingyun Lu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Integrated Traditional Chinese and Western medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lu Liu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cui Wang
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Xie
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Li
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Tian
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Kim DY, Ko SH. Common Regulators of Lipid Metabolism and Bone Marrow Adiposity in Postmenopausal Women. Pharmaceuticals (Basel) 2023; 16:322. [PMID: 37259464 PMCID: PMC9967016 DOI: 10.3390/ph16020322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 09/13/2024] Open
Abstract
A variety of metabolic disorders are associated with a decrease in estradiol (E2) during natural or surgical menopause. Postmenopausal women are prone to excessive fat accumulation in skeletal muscle and adipose tissue due to the loss of E2 via abnormalities in lipid metabolism and serum lipid levels. In skeletal muscle and adipose tissue, genes related to energy metabolism and fatty acid oxidation, such as those encoding peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) and estrogen-related receptor alpha (ERRα), are downregulated, leading to increased fat synthesis and lipid metabolite accumulation. The same genes regulate lipid metabolism abnormalities in the bone marrow. In this review, abnormalities in lipid metabolism caused by E2 deficiency were investigated, with a focus on genes able to simultaneously regulate not only skeletal muscle and adipose tissue but also bone metabolism (e.g., genes encoding PGC-1α and ERRα). In addition, the mechanisms through which mesenchymal stem cells lead to adipocyte differentiation in the bone marrow as well as metabolic processes related to bone marrow adiposity, bone loss, and osteoporosis were evaluated, focusing on the loss of E2 and lipid metabolic alterations. The work reviewed here suggests that genes underlying lipid metabolism and bone marrow adiposity are candidate therapeutic targets for bone loss and osteoporosis in postmenopausal women.
Collapse
Affiliation(s)
- Dae-Yong Kim
- CEO, N- BIOTEK, Inc., 402-803, Technopark, 655, Pyeongcheon-ro, Bucheon-si 14502, Gyeonggi-do, Republic of Korea
| | - Seong-Hee Ko
- Regenerative Medicine Research Team, N- BIOTEK, Inc., 104-706, Technopark Ssangyong 3Cha, 397, Seokcheon-ro, Bucheon-si 14449, Gyeonggi-do, Republic of Korea
| |
Collapse
|
10
|
Beekman KM, Duque G, Corsi A, Tencerova M, Bisschop PH, Paccou J. Osteoporosis and Bone Marrow Adipose Tissue. Curr Osteoporos Rep 2023; 21:45-55. [PMID: 36534306 DOI: 10.1007/s11914-022-00768-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW This review focuses on the recent findings regarding bone marrow adipose tissue (BMAT) concerning bone health. We summarize the variations in BMAT in relation to age, sex, and skeletal sites, and provide an update on noninvasive imaging techniques to quantify human BMAT. Next, we discuss the role of BMAT in patients with osteoporosis and interventions that affect BMAT. RECENT FINDINGS There are wide individual variations with region-specific fluctuation and age- and gender-specific differences in BMAT content and composition. The Bone Marrow Adiposity Society (BMAS) recommendations aim to standardize imaging protocols to increase comparability across studies and sites. Water-fat imaging (WFI) seems an accurate and efficient alternative for spectroscopy (1H-MRS). Most studies indicate that greater BMAT is associated with lower bone mineral density (BMD) and a higher prevalence of vertebral fractures. The proton density fat fraction (PDFF) and changes in lipid composition have been associated with an increased risk of fractures independently of BMD. Therefore, PDFF and lipid composition could potentially be future imaging biomarkers for assessing fracture risk. Evidence of the inhibitory effect of osteoporosis treatments on BMAT is still limited to a few randomized controlled trials. Moreover, results from the FRAME biopsy sub-study highlight contradictory findings on the effect of the sclerostin antibody romosozumab on BMAT. Further understanding of the role(s) of BMAT will provide insight into the pathogenesis of osteoporosis and may lead to targeted preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Kerensa M Beekman
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Gustavo Duque
- Department of Medicine and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Michaela Tencerova
- Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Peter H Bisschop
- Department of Endocrinology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Julien Paccou
- Department of Rheumatology, MABLaB ULR 4490, CHU Lille, University Lille, Lille, France.
| |
Collapse
|
11
|
Abstract
Changes in bone architecture and metabolism with aging increase the likelihood of osteoporosis and fracture. Age-onset osteoporosis is multifactorial, with contributory extrinsic and intrinsic factors including certain medical problems, specific prescription drugs, estrogen loss, secondary hyperparathyroidism, microenvironmental and cellular alterations in bone tissue, and mechanical unloading or immobilization. At the histological level, there are changes in trabecular and cortical bone as well as marrow cellularity, lineage switching of mesenchymal stem cells to an adipogenic fate, inadequate transduction of signals during skeletal loading, and predisposition toward senescent cell accumulation with production of a senescence-associated secretory phenotype. Cumulatively, these changes result in bone remodeling abnormalities that over time cause net bone loss typically seen in older adults. Age-related osteoporosis is a geriatric syndrome due to the multiple etiologies that converge upon the skeleton to produce the ultimate phenotypic changes that manifest as bone fragility. Bone tissue is dynamic but with tendencies toward poor osteoblastic bone formation and relative osteoclastic bone resorption with aging. Interactions with other aging physiologic systems, such as muscle, may also confer detrimental effects on the aging skeleton. Conversely, individuals who maintain their BMD experience a lower risk of fractures, disability, and mortality, suggesting that this phenotype may be a marker of successful aging. © 2023 American Physiological Society. Compr Physiol 13:4355-4386, 2023.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of Medicine, Divisions of Geriatric Medicine and Gerontology, Endocrinology, and Hospital Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,The Department of Physiology and Biomedical Engineering, and the Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
12
|
Sarkis R, Burri O, Royer-Chardon C, Schyrr F, Blum S, Costanza M, Cherix S, Piazzon N, Barcena C, Bisig B, Nardi V, Sarro R, Ambrosini G, Weigert M, Spertini O, Blum S, Deplancke B, Seitz A, de Leval L, Naveiras O. MarrowQuant 2.0: A Digital Pathology Workflow Assisting Bone Marrow Evaluation in Experimental and Clinical Hematology. Mod Pathol 2023; 36:100088. [PMID: 36788087 DOI: 10.1016/j.modpat.2022.100088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/22/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023]
Abstract
Bone marrow (BM) cellularity assessment is a crucial step in the evaluation of BM trephine biopsies for hematologic and nonhematologic disorders. Clinical assessment is based on a semiquantitative visual estimation of the hematopoietic and adipocytic components by hematopathologists, which does not provide quantitative information on other stromal compartments. In this study, we developed and validated MarrowQuant 2.0, an efficient, user-friendly digital hematopathology workflow integrated within QuPath software, which serves as BM quantifier for 5 mutually exclusive compartments (bone, hematopoietic, adipocytic, and interstitial/microvasculature areas and other) and derives the cellularity of human BM trephine biopsies. Instance segmentation of individual adipocytes is realized through the adaptation of the machine-learning-based algorithm StarDist. We calculated BM compartments and adipocyte size distributions of hematoxylin and eosin images obtained from 250 bone specimens, from control subjects and patients with acute myeloid leukemia or myelodysplastic syndrome, at diagnosis and follow-up, and measured the agreement of cellularity estimates by MarrowQuant 2.0 against visual scores from 4 hematopathologists. The algorithm was capable of robust BM compartment segmentation with an average mask accuracy of 86%, maximal for bone (99%), hematopoietic (92%), and adipocyte (98%) areas. MarrowQuant 2.0 cellularity score and hematopathologist estimations were highly correlated (R2 = 0.92-0.98, intraclass correlation coefficient [ICC] = 0.98; interobserver ICC = 0.96). BM compartment segmentation quantitatively confirmed the reciprocity of the hematopoietic and adipocytic compartments. MarrowQuant 2.0 performance was additionally tested for cellularity assessment of specimens prospectively collected from clinical routine diagnosis. After special consideration for the choice of the cellularity equation in specimens with expanded stroma, performance was similar in this setting (R2 = 0.86, n = 42). Thus, we conclude that these validation experiments establish MarrowQuant 2.0 as a reliable tool for BM cellularity assessment. We expect this workflow will serve as a clinical research tool to explore novel biomarkers related to BM stromal components and may contribute to further validation of future digitalized diagnostic hematopathology workstreams.
Collapse
Affiliation(s)
- Rita Sarkis
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering & ISREC, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland; Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Olivier Burri
- BioImaging and Optics Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Claire Royer-Chardon
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Frédérica Schyrr
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering & ISREC, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sophie Blum
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering & ISREC, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Mariangela Costanza
- Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Stephane Cherix
- Department of Orthopaedics and Traumatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nathalie Piazzon
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Carmen Barcena
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland; Department of Pathology, Hospital 12 de Octubre, Madrid, Spain
| | - Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Rossella Sarro
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland; Institute of Pathology, Ente Ospedaliero Cantonale (EOC), Locarno, Switzerland
| | - Giovanna Ambrosini
- Bioinformatics Competence Center (BICC), UNIL/EPFL Lausanne, Switzerland
| | - Martin Weigert
- Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Olivier Spertini
- Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Sabine Blum
- Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Arne Seitz
- BioImaging and Optics Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering & ISREC, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
13
|
Douglass A, Dattilo M, Feola AJ. Evidence for Menopause as a Sex-Specific Risk Factor for Glaucoma. Cell Mol Neurobiol 2023; 43:79-97. [PMID: 34981287 PMCID: PMC9250947 DOI: 10.1007/s10571-021-01179-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/03/2021] [Indexed: 01/07/2023]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by progressive loss of visual function and retinal ganglion cells (RGC). Current epidemiological, clinical, and basic science evidence suggest that estrogen plays a role in the aging of the optic nerve. Menopause, a major biological life event affecting all women, coincides with a decrease in circulating sex hormones, such as estrogen. While 59% of the glaucomatous population are females, sex is not considered a risk factor for developing glaucoma. In this review, we explore whether menopause is a sex-specific risk factor for glaucoma. First, we investigate how menopause is defined as a sex-specific risk factor for other pathologies, including cardiovascular disease, osteoarthritis, and bone health. Next, we discuss clinical evidence that highlights the potential role of menopause in glaucoma. We also highlight preclinical studies that demonstrate larger vision and RGC loss following surgical menopause and how estrogen is protective in models of RGC injury. Lastly, we explore how surgical menopause and estrogen signaling are related to risk factors associated with developing glaucoma (e.g., intraocular pressure, aqueous outflow resistance, and ocular biomechanics). We hypothesize that menopause potentially sets the stage to develop glaucoma and therefore is a sex-specific risk factor for this disease.
Collapse
Affiliation(s)
- Amber Douglass
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA
| | - Michael Dattilo
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, B2503, Clinic B Building, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
- Department of Ophthalmology, Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA
- Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrew J Feola
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA.
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, B2503, Clinic B Building, 1365B Clifton Road NE, Atlanta, GA, 30322, USA.
- Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
14
|
Lin X, Xu F, Zhang KW, Qiu WX, Zhang H, Hao Q, Li M, Deng XN, Tian Y, Chen ZH, Qian AR. Acacetin Prevents Bone Loss by Disrupting Osteoclast Formation and Promoting Type H Vessel Formation in Ovariectomy-Induced Osteoporosis. Front Cell Dev Biol 2022; 10:796227. [PMID: 35517504 PMCID: PMC9062130 DOI: 10.3389/fcell.2022.796227] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/01/2022] [Indexed: 12/20/2022] Open
Abstract
Osteoporosis, characterized by the destruction of bone resorption and bone formation, is a serious disease that endangers human health. Osteoporosis prevention and treatment has become one of the important research contents in the field of medicine. Acacetin, a natural flavonoid compound, could promote osteoblast differentiation, and inhibit osteoclast formation in vitro. However, the mechanisms of acacetin on osteoclast differentiation and type H vessel formation, as well as the effect of preventing bone loss, remain unclear. Here, we firstly used primary bone marrow derived macrophages (BMMs), endothelial progenitor cells (EPCs), and ovariectomized (OVX) mice to explore the function of acacetin on bone remodeling and H type vessel formation. In this study, we found that acacetin inhibits osteoclast formation and bone resorption of BMMs induced by the macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) in a concentration of 20 μM without exerting cytotoxic effects. It was accompanied by downregulation of osteoclast differentiation marker genes (Ctsk, Acp5, and Mmp9) and cell fusion genes (CD9, CD47, Atp6v0d2, Dc-stamp, and Oc-stamp). Moreover, acacetin disrupted actin ring formation and extracellular acidification in osteoclasts. Mechanistic analysis revealed that acacetin not only inhibits the expression of the major transcription factor NFATc1 and NF-κB during RANKL-induced osteoclast formation, but also suppresses RANKL-induced the phosphorylation of Akt, GSK3β, IκBα, and p65. Additionally, acacetin enhanced the ability of M-CSF and RANKL-stimulated BMMs to promote angiogenesis and migration of EPCs. We further established that, in vivo, acacetin increased trabecular bone mass, decreased the number of osteoclasts, and showed more type H vessels in OVX mice. These data demonstrate that acacetin prevents OVX-induced bone loss in mice through inhibition of osteoclast function and promotion of type H vessel formation via Akt/GSK3β and NF-κB signalling pathway, suggesting that acacetin may be a novel therapeutic agent for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiao Lin
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Fang Xu
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Ke-Wen Zhang
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Wu-Xia Qiu
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Hui Zhang
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Xiao-Ni Deng
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Ye Tian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Ye Tian, ; Zhi-Hao Chen, ; Ai-Rong Qian,
| | - Zhi-Hao Chen
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Ye Tian, ; Zhi-Hao Chen, ; Ai-Rong Qian,
| | - Ai-Rong Qian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Ye Tian, ; Zhi-Hao Chen, ; Ai-Rong Qian,
| |
Collapse
|
15
|
Londraville RL, Tuttle M, Liu Q, Andronowski JM. Endospanin Is a Candidate for Regulating Leptin Sensitivity. Front Physiol 2022; 12:786299. [PMID: 35069248 PMCID: PMC8777038 DOI: 10.3389/fphys.2021.786299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
The hypothesis advanced is that endospanin, a highly conserved vesicle traffic protein in vertebrates, regulates leptin sensitivity in bone signaling. The effects of leptin on bones are well-studied but without consensus on whether the increases in leptin signaling stimulate bone gain or loss. The bone response may depend on leptin sensitivity, and endospanin is an established modulator of leptin sensitivity. An argument is advanced to develop zebrafish models for specific leptin signaling pathways. Zebrafish have well-developed molecular tools (e.g., CRISPR) and the advantage of non-destructive sampling of bones in the form of scales. Using these tools, experiments are described to substantiate the role of endospanin in zebrafish bone dynamics.
Collapse
Affiliation(s)
- Richard L. Londraville
- Program in Integrated Bioscience, Department of Biology, University of Akron, Akron, OH, United States
| | - Matthew Tuttle
- Program in Integrated Bioscience, Department of Biology, University of Akron, Akron, OH, United States
| | - Qin Liu
- Program in Integrated Bioscience, Department of Biology, University of Akron, Akron, OH, United States
| | - Janna M. Andronowski
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. Johns, NL, Canada
| |
Collapse
|
16
|
Batoon L, Millard SM, Raggatt LJ, Wu AC, Kaur S, Sun LWH, Williams K, Sandrock C, Ng PY, Irvine KM, Bartnikowski M, Glatt V, Pavlos NJ, Pettit AR. Osteal macrophages support osteoclast-mediated resorption and contribute to bone pathology in a postmenopausal osteoporosis mouse model. J Bone Miner Res 2021; 36:2214-2228. [PMID: 34278602 DOI: 10.1002/jbmr.4413] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/29/2021] [Accepted: 07/14/2021] [Indexed: 11/08/2022]
Abstract
Osteal macrophages (osteomacs) support osteoblast function and promote bone anabolism, but their contribution to osteoporosis has not been explored. Although mouse ovariectomy (OVX) models have been repeatedly used, variation in strain, experimental design and assessment modalities have contributed to no single model being confirmed as comprehensively replicating the full gamut of osteoporosis pathological manifestations. We validated an OVX model in adult C3H/HeJ mice and demonstrated that it presents with human postmenopausal osteoporosis features with reduced bone volume in axial and appendicular bone and bone loss in both trabecular and cortical bone including increased cortical porosity. Bone loss was associated with increased osteoclasts on trabecular and endocortical bone and decreased osteoblasts on trabecular bone. Importantly, this OVX model was characterized by delayed fracture healing. Using this validated model, we demonstrated that osteomacs are increased post-OVX on both trabecular and endocortical bone. Dual F4/80 (pan-macrophage marker) and tartrate-resistant acid phosphatase (TRAP) staining revealed osteomacs frequently located near TRAP+ osteoclasts and contained TRAP+ intracellular vesicles. Using an in vivo inducible macrophage depletion model that does not simultaneously deplete osteoclasts, we observed that osteomac loss was associated with elevated extracellular TRAP in bone marrow interstitium and increased serum TRAP. Using in vitro high-resolution confocal imaging of mixed osteoclast-macrophage cultures on bone substrate, we observed macrophages juxtaposed to osteoclast basolateral functional secretory domains scavenging degraded bone byproducts. These data demonstrate a role for osteomacs in supporting osteoclastic bone resorption through phagocytosis and sequestration of resorption byproducts. Overall, our data expose a novel role for osteomacs in supporting osteoclast function and provide the first evidence of their involvement in osteoporosis pathogenesis. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Lena Batoon
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Susan M Millard
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Liza J Raggatt
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Andy C Wu
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Simranpreet Kaur
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Lucas W H Sun
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Kyle Williams
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Cheyenne Sandrock
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Pei Ying Ng
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Katharine M Irvine
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Michal Bartnikowski
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Vaida Glatt
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,Orthopaedic Surgery Department, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Nathan J Pavlos
- Bone Biology and Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Allison R Pettit
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| |
Collapse
|
17
|
Beekman KM, Akkerman EM, Streekstra GJ, Veldhuis‐Vlug AG, Acherman Y, Gerdes VE, den Heijer M, Maas M, Bravenboer N, Bisschop PH. The Effect of Roux-en-Y Gastric Bypass on Bone Marrow Adipose Tissue and Bone Mineral Density in Postmenopausal, Nondiabetic Women. Obesity (Silver Spring) 2021; 29:1120-1127. [PMID: 33951317 PMCID: PMC8359834 DOI: 10.1002/oby.23171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVES This study aimed to determine the effect of bariatric surgery-induced weight loss on bone marrow adipose tissue (BMAT) and bone mineral density (BMD) in postmenopausal, nondiabetic women. METHODS A total of 14 postmenopausal, nondiabetic women with obesity who were scheduled for laparoscopic Roux-en-Y gastric bypass surgery (RYGB) were included in this study. Vertebral bone marrow fat signal fraction was determined by quantitative chemical shift magnetic resonance imaging, and vertebral volumetric BMD (vBMD) was determined by quantitative computed tomography before surgery and 3 and 12 months after surgery. Data were analyzed by linear mixed model. RESULTS Body weight [mean (SD)] decreased after surgery from 108 (13) kg at baseline to 89 (12) kg at 3 months and 74 (11) kg at 12 months (P < 0.001). BMAT decreased after surgery from 51% (8%) at baseline to 50% (8%) at 3 months and 46% (7%) at 12 months (P = 0.004). vBMD decreased after surgery from 101 (26) mg/cm3 at baseline to 94 (28) mg/cm3 at 3 months (P = 0.003) and 94 (28) mg/cm3 at 12 months (P = 0.035). Changes in BMAT and vBMD were not correlated (ρ = -0.10 and P = 0.75). Calcium and vitamin D concentrations did not change after surgery. CONCLUSIONS RYGB decreases both BMAT (after 12 months) and vBMD (both after 3 months and 12 months) in postmenopausal, nondiabetic women. Changes in BMAT and vBMD were not correlated. These findings suggest that BMAT does not contribute to bone loss following RYGB.
Collapse
Affiliation(s)
- Kerensa M. Beekman
- Department of Radiology and Nuclear MedicineAmsterdam Movement SciencesAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
- Department of EndocrinologyAmsterdam Movement SciencesAmsterdam University Medical CenterVrije University, AmsterdamAmsterdamthe Netherlands
| | - Erik M. Akkerman
- Department of Radiology and Nuclear MedicineAmsterdam Movement SciencesAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | - Geert J. Streekstra
- Department of Radiology and Nuclear MedicineAmsterdam Movement SciencesAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
- Department of Biomedical Engineering and PhysicsAmsterdam Movement SciencesAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | - Annegreet G. Veldhuis‐Vlug
- Department of Internal MedicineJan van Goyen Medical Center/Onze Lieve Vrouwe GasthuisAmsterdamthe Netherlands
- Department of EndocrinologyLeiden University Medical CenterLeidenthe Netherlands
| | - Yair Acherman
- Department of SurgerySpaarne GasthuisHaarlemthe Netherlands
| | - Victor E. Gerdes
- Department of Vascular MedicineAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | - Martin den Heijer
- Department of EndocrinologyAmsterdam Movement SciencesAmsterdam University Medical CenterVrije University, AmsterdamAmsterdamthe Netherlands
| | - Mario Maas
- Department of Radiology and Nuclear MedicineAmsterdam Movement SciencesAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | - Nathalie Bravenboer
- Department of EndocrinologyLeiden University Medical CenterLeidenthe Netherlands
- Department of Clinical ChemistryResearch Laboratory Bone and Calcium MetabolismAmsterdam Movement SciencesAmsterdam University Medical CenterVrije University, AmsterdamAmsterdamthe Netherlands
| | - Peter H. Bisschop
- Department of EndocrinologyAmsterdam Movement SciencesAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
18
|
Pignolo RJ, Law SF, Chandra A. Bone Aging, Cellular Senescence, and Osteoporosis. JBMR Plus 2021; 5:e10488. [PMID: 33869998 PMCID: PMC8046105 DOI: 10.1002/jbm4.10488] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
Changes in aging bone that lead to osteoporosis are mediated at multiple levels, including hormonal alterations, skeletal unloading, and accumulation of senescent cells. This pathological interplay is superimposed upon medical conditions, potentially bone-wasting medications, modifiable and unmodifiable personal risk factors, and genetic predisposition that accelerate bone loss with aging. In this study, the focus is on bone hemostasis and its dysregulation with aging. The major physiological changes with aging in bone and the role of cellular senescence in contributing to age-related osteoporosis are summarized. The aspects of bone aging are reviewed including remodeling deficits, uncoupling phenomena, inducers of cellular senescence related to bone aging, roles of the senescence-associated secretory phenotype, radiation-induced bone loss as a model for bone aging, and the accumulation of senescent cells in the bone microenvironment as a predominant mechanism for age-related osteoporosis. The study also addresses the rationale and potential for therapeutic interventions based on the clearance of senescent cells or suppression of the senescence-associated secretory phenotype. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of MedicineMayo ClinicRochesterMNUSA
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMNUSA
| | - Susan F Law
- Department of MedicineMayo ClinicRochesterMNUSA
| | - Abhishek Chandra
- Department of MedicineMayo ClinicRochesterMNUSA
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMNUSA
| |
Collapse
|
19
|
Lee H, Kim MH, Choi LY, Yang WM. Ameliorative effects of Osteo-F, a newly developed herbal formula, on osteoporosis via activation of bone formation. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113590. [PMID: 33212177 DOI: 10.1016/j.jep.2020.113590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The fruit of Schizandra chinensis and Lycium chinense, and the root of Eucommia ulmoides, components of Osteo-F, has traditionally been used for treating bone diseases in Korean Medicine. AIM OF THE STUDY The exact role and underlying mechanism of Osteo-F herbal formula on bone formation in osteoporosis was investigated in the present study. MATERIALS AND METHODS OVX mice were treated with 0.9, 9 and 90 mg/kg of Osteo-F for 4 weeks. Bone tissues including fourth to sixth lumbar vertebrae (LV) and femur were collected to analyze the bone mineral density (BMD). In addition, serum biomarkers were estimated by enzyme-linked immunosorbent assay. The expressions of collagen, BMP-2 and osteopontin were determined in tibia to clarify the bone anabolic effects of Osteo-F in osteoporosis. RESULTS The levels of BMD in both of fourth to sixth LV and femur were significantly increased by Osteo-F treatment in OVX mice. Bone mineral content (BMC) was also elevated in Osteo-F-treated LV and femoral bone tissues. In addition, serum osteocalcin was markedly increased by Osteo-F in osteoporotic mice. Serum ALP and bALP levels were neutralized in Osteo-F 90 mg/kg-administered mice. Furthermore, Osteo-F treatment dramatically increased the mRNA expressions of collagen type I, BMP-2 and OPN in tibial bone specimens. CONCLUSIONS Osteo-F ameliorated bone loss by increasing bone forming molecules including BMP-2 and OPN in osteoporosis. Osteo-F, a newly developed herbal formula, may be an alternative material for the management of osteoporosis with bone anabolic effects.
Collapse
Affiliation(s)
- Haesu Lee
- BOINBIO Convergence Co., Ltd, Seoulbioherb, 117-3 Hoegi-ro, Dongdaemun-gu, Seoul, 02455, Republic of Korea; Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Mi Hye Kim
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - La Yoon Choi
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Woong Mo Yang
- Department of Convergence Korean Medical Science, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
20
|
Cici D, Corrado A, Rotondo C, Colia R, Cantatore FP. Adipokines and Chronic Rheumatic Diseases: from Inflammation to Bone Involvement. Clin Rev Bone Miner Metab 2021. [DOI: 10.1007/s12018-021-09275-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractBesides its well-known role as energy storage tissue, adipose tissue is a biologically active tissue that can also be considered as an endocrine organ, as it is able to secrete adipokines. These bioactive factors, similar in structure to cytokines, are involved in several physiological and pathological conditions, such as glucose homeostasis, angiogenesis, blood pressure regulation, control of food intake, and also inflammation and bone homeostasis via endocrine, paracrine, and autocrine mechanisms. Given their pleiotropic functions, the role of adipokines has been evaluated in chronic rheumatic osteoarticular inflammatory diseases, particularly focusing on their effects on inflammatory and immune response and on bone alterations. Indeed, these diseases are characterized by different bone complications, such as local and systemic bone loss and new bone formation. The aim of this review is to summarize the role of adipokines in rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, osteoarthritis, and osteoporosis, especially considering their role in the pathogenesis of bone complications typical of these conditions.
Collapse
|
21
|
Li L, Zhou X, Zhang JT, Liu AF, Zhang C, Han JC, Zhang XQ, Wu S, Zhang XY, Lv FQ. Exosomal miR-186 derived from BMSCs promote osteogenesis through hippo signaling pathway in postmenopausal osteoporosis. J Orthop Surg Res 2021; 16:23. [PMID: 33413543 PMCID: PMC7791800 DOI: 10.1186/s13018-020-02160-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Postmenopausal osteoporosis (PMO) that results from estrogen withdrawal is the most common primary osteoporosis among older women. However, little is known about the mechanism of PMO, and effective treatment of PMO is limited. METHODS We used real-time polymerase chain reaction (qPCR), Western blotting, and RNA pull down to investigate the relationship between miR-186 and MOB Kinase Activator 1A (Mob1). Also, we investigated the effect of exosome in osteogenesis using alkaline phosphatase (ALP) staining. And hematoxylin eosin (HE) staining was used to verify the osteogenesis in PMO model. RESULTS Exosomal miR-186 plays an important role in bone formation. The results of miRNA-seq and q-PCR showed that miR-186 was upregulated in a PMO + Exo treatment group. Results of RNA-pull down and luciferase reporter assays verified interactions between miR-186 and Mob1. We also verified the Hippo signaling pathway plays an important role in osteogenesis. CONCLUSIONS We concluded that exosomes derived from human bone marrow mesenchymal stem cells (hBMSCs) can transfer miR-186 to promote osteogenesis in ovariectomy (OVX) rats through the Hippo signaling pathway.
Collapse
Affiliation(s)
- Lu Li
- Department of Acupuncture, Tianjin Nankai Hospital, Tianjin, 300100, People's Republic of China
| | - Xin Zhou
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China
| | - Jun-Tao Zhang
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China
| | - Ai-Feng Liu
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China
| | - Chao Zhang
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China
| | - Jin-Chang Han
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China
| | - Xiao-Qing Zhang
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China
| | - Si Wu
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China
| | - Xiao-Yu Zhang
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, People's Republic of China.
| | - Fu-Quan Lv
- Department of Acupuncture, Tianjin Nankai Hospital, Tianjin, 300100, People's Republic of China.
| |
Collapse
|
22
|
Lee JY, Yang JY, Kim SW. Bone Lining Cells Could Be Sources of Bone Marrow Adipocytes. Front Endocrinol (Lausanne) 2021; 12:766254. [PMID: 34925236 PMCID: PMC8678613 DOI: 10.3389/fendo.2021.766254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recently, lineage-tracing studies demonstrated that parathyroid hormone and anti-sclerostin antibody (Scl-Ab) can convert bone lining cells (BLCs) into active osteoblasts. However, BLCs might also be differentiated into other lineages. Here we investigated whether BLCs could differentiate into bone marrow adipocytes (BMAds) and whether Scl-Ab could suppress this process. METHODS Dmp1-CreERt2:mTmG mice were injected with 0.5 mg of 4-hydroxytamoxifen once weekly from postnatal week 4 to week 8. The mice were treated with either vehicle or rosiglitazone for 8 weeks (weeks 12-20). Moreover, they were administered either vehicle or Scl-Ab (50 mg/kg) twice weekly for 4 weeks (weeks 16-20, N = 4-6/group). We chased the GFP+ cells from the endosteal surface to the bone marrow (BM) of the femur. Using immunohistochemical staining, the numbers of perilipin+ or GFP+/perilipin double+ cells in the BM were quantified. In addition, serum N-terminal propeptide of type I procollagen (P1NP) levels were measured at each time point, and bone mass was analyzed at 20 weeks using micro-computed tomography. RESULTS Scl-Ab administration significantly reversed the decreases in bone parameters induced by rosiglitazone. Plump GFP+ cells, presumably active osteoblasts, and extremely flat GFP+ cells, presumably BLCs, were present on the endosteal surface of the femur at 8 and 12 weeks, respectively, in line with prior findings. When we chased the GFP+ cells, rosiglitazone significantly increased the number of GFP/perilipin double+ BMAds compared to the effects of the vehicle (P < 0.001), and overlapping Scl-Ab administration decreased the number of GFP/perilipin double + BMAd compared to rosiglitazone alone (P < 0.001). In addition, we found that osteoblast lineage cells such as BLCs might express PPARγ on immunohistochemical staining. When rosiglitazone was administered to Rip-Cre:mTmG mice, GFP+ cells were not present on the endosteal surface or in the BM of the femur; however, they were present in the pancreas. CONCLUSION BLCs could be sources of BMAds, and rosiglitazone could stimulate the differentiation of osteoblast lineage cells into BMAds. Suppression of the differentiation of osteoblast lineage cells into BMAds might contribute to anabolic effects resulting from the pharmacologic inhibition of sclerostin.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Research and Experiment, Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| | - Jae-Yeon Yang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Research and Experiment, Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea
| | - Sang Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Division of Endocrinology and Metabolism, Seoul Metropolitan Government Boramae Medical Center, Seoul, South Korea
- *Correspondence: Sang Wan Kim,
| |
Collapse
|
23
|
Omagari D, Hayatsu M, Yamamoto K, Kobayashi M, Tsukano N, Nameta M, Mikami Y. Gap junction with MLO-A5 osteoblast-like cell line induces ALP and BSP transcription of 3T3-L1 pre-adipocyte like cell line via Hspb1 while retaining adipogenic differentiation ability. Bone 2020; 141:115596. [PMID: 32814124 DOI: 10.1016/j.bone.2020.115596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/09/2022]
Abstract
In bone tissues, gap junctions form direct links between the cytoplasm of an osteocyte and another adjacent osteocyte or osteoblast, which underlie both bone formation and bone resorption. We have previously demonstrated that alkaline phosphatase (ALP) and bone sialoprotein (BSP), which are osteoblast markers, were induced in mesenchymal stem cells (MSCs) co-cultured with osteoblast-like cell line. However, the molecular mechanism of this process has not been fully addressed. Furthermore, few advances have been made toward elucidating the communication networks that link the status of committed cells such as (pre-) adipocytes that differentiated from MSCs as well as osteoblasts. Therefore, the objective of the present study was to investigate the mechanism underlying the communication network between pre-adipocytes and osteoblasts. We evaluated the effect of co-culture with osteoblast on the cell status of pre-adipocytes using murine osteoblast-like cell line, MLO-A5, and pre-adipocyte-like cell line, 3T3-L1, respectively. The results presented here demonstrated that osteoblasts and pre-adipocytes communicate via gap junctions, and the ensuing drastic increase in ALP and BSP transcription in co-cultured pre-adipocytes was induced, at least partly, via heat shock protein family B member 1 (Hspb1). In addition, terminal differentiation into adipocytes was suppressed in pre-adipocytes during co-culture with osteoblast without loss of adipogenic differentiation ability. Interestingly, after co-culture with osteoblasts, isolated co-cultured pre-adipocytes were able to differentiate to adipocytes as well as original pre-adipocytes. These results suggest that gap junctional communication with osteoblasts suppressed adipogenic differentiation of pre-adipocytes without loss of adipogenic differentiation ability.
Collapse
Affiliation(s)
- Daisuke Omagari
- Department of Pathology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Manabu Hayatsu
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata-shi, Niigata 951-8122, Japan
| | - Kiyofumi Yamamoto
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Naruchika Tsukano
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata-shi, Niigata 951-8122, Japan
| | - Masaaki Nameta
- Electron Microscope Core Facility, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata-shi, Niigata 951-8122, Japan
| | - Yoshikazu Mikami
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata-shi, Niigata 951-8122, Japan.
| |
Collapse
|
24
|
A Review of the Action of Magnesium on Several Processes Involved in the Modulation of Hematopoiesis. Int J Mol Sci 2020; 21:ijms21197084. [PMID: 32992944 PMCID: PMC7582682 DOI: 10.3390/ijms21197084] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022] Open
Abstract
Magnesium (Mg2+) is an essential mineral for the functioning and maintenance of the body. Disturbances in Mg2+ intracellular homeostasis result in cell-membrane modification, an increase in oxidative stress, alteration in the proliferation mechanism, differentiation, and apoptosis. Mg2+ deficiency often results in inflammation, with activation of inflammatory pathways and increased production of proinflammatory cytokines by immune cells. Immune cells and others that make up the blood system are from hematopoietic tissue in the bone marrow. The hematopoietic tissue is a tissue with high indices of renovation, and Mg2+ has a pivotal role in the cell replication process, as well as DNA and RNA synthesis. However, the impact of the intra- and extracellular disturbance of Mg2+ homeostasis on the hematopoietic tissue is little explored. This review deals specifically with the physiological requirements of Mg2+ on hematopoiesis, showing various studies related to the physiological requirements and the effects of deficiency or excess of this mineral on the hematopoiesis regulation, as well as on the specific process of erythropoiesis, granulopoiesis, lymphopoiesis, and thrombopoiesis. The literature selected includes studies in vitro, in animal models, and in humans, giving details about the impact that alterations of Mg2+ homeostasis can have on hematopoietic cells and hematopoietic tissue.
Collapse
|
25
|
Murata Y, Uchida S, Utsunomiya H, Hatakeyama A, Nakashima H, Mori T, Yamanaka Y, Tsukamoto M, Sekiya I, Huard J, Philippon MJ, Sakai A. Differentiation Potential of Synovial Mesenchymal Stem Cells Isolated From Hip Joints Affected by Femoroacetabular Impingement Syndrome Versus Osteoarthritis. Arthroscopy 2020; 36:2122-2133. [PMID: 32259644 DOI: 10.1016/j.arthro.2020.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/06/2020] [Accepted: 03/03/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE To establish the characteristics of synovium-derived mesenchymal stem cells (MSCs) from the hip joints of patients with femoroacetabular impingement syndrome (FAIS) and osteoarthritis (OA), particularly their proliferation and differentiation potentials. We further investigated their functional differences. METHODS Synovium samples were harvested from 21 patients with FAIS who underwent hip arthroscopic surgery and from 14 patients with OA who underwent total hip arthroplasty. The MSC number, colony-forming units, cell viability, and differentiation potential were compared. Real-time polymerase chain reaction assessed the differentiation potential into adipose, bone, and cartilage tissues. RESULTS The number of colonies at a density of 104 at passage 0 from OA synovium was significantly greater than that from FAIS synovium (P < .01). However, their proliferation and viability were significantly lower than those of FAIS synovium cells (P = .0495). The expression of lipoprotein lipase mRNA in OA synovium cells was greater than that in FAIS synovium cells (P < .01). Meanwhile, the fraction of colonies positive for von Kossa and alkaline phosphatase staining, as well as the level of bone gamma-carboxyglutamate protein expression in OA synovium cells, were greater than those in FAIS synovium cells (P < .01). In chondrogenic pellet culture experiments, the expression of COL10A1 mRNA was lower in OA synovium than in FAIS synovium (P < .01). CONCLUSIONS Synovial MSCs from patients with OA had greater colony numbers but less viability and proliferative potential. They also showed greater osteogenic and adipogenic potentials, whereas those from patients with FAIS showed greater chondrogenic potential. CLINICAL RELEVANCE MSCs from patients with FAIS exhibited good potential as cell sources for stem cell therapy in case of cartilage damage in the hip joint.
Collapse
Affiliation(s)
- Yoichi Murata
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Soshi Uchida
- Department of Orthopaedic Surgery and Sports Medicine, Wakamatsu Hospital of University of Occupational and Environmental Health, Kitakyushu, Japan.
| | - Hajime Utsunomiya
- Department of Orthopaedic Surgery and Sports Medicine, Wakamatsu Hospital of University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Akihisa Hatakeyama
- Department of Orthopaedic Surgery and Sports Medicine, Wakamatsu Hospital of University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hirotaka Nakashima
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Toshiharu Mori
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiaki Yamanaka
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Manabu Tsukamoto
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Johnny Huard
- The Steadman Philippon Research Institute and the Steadman Clinic, Vail, Colorado, U.S.A
| | - Marc J Philippon
- The Steadman Philippon Research Institute and the Steadman Clinic, Vail, Colorado, U.S.A
| | - Akinori Sakai
- Department of Orthopaedic Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
26
|
Electrochemical anticancer drug sensor for determination of raloxifene in the presence of tamoxifen using graphene-CuO-polypyrrole nanocomposite structure modified pencil graphite electrode: Theoretical and experimental investigation. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2020.113314] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
27
|
Li J, Chen X, Lu L, Yu X. The relationship between bone marrow adipose tissue and bone metabolism in postmenopausal osteoporosis. Cytokine Growth Factor Rev 2020; 52:88-98. [PMID: 32081538 DOI: 10.1016/j.cytogfr.2020.02.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 02/06/2023]
Abstract
Postmenopausal osteoporosis (PMOP) is a prevalent skeletal disorder associated with menopause-related estrogen withdrawal. PMOP is characterized by low bone mass, deterioration of the skeletal microarchitecture, and subsequent increased susceptibility to fragility fractures, thus contributing to disability and mortality. Accumulating evidence indicates that abnormal expansion of marrow adipose tissue (MAT) plays a crucial role in the onset and progression of PMOP, in part because both bone marrow adipocytes and osteoblasts share a common ancestor lineage. The cohabitation of MAT adipocytes, mesenchymal stromal cells, hematopoietic cells, osteoblasts and osteoclasts in the bone marrow creates a microenvironment that permits adipocytes to act directly on other cell types in the marrow. Furthermore, MAT, which is recognized as an endocrine organ, regulates bone remodeling through the secretion of adipokines and cytokines. Although an enhanced MAT volume is linked to low bone mass and fractures in PMOP, the detailed interactions between MAT and bone metabolism remain largely unknown. In this review, we examine the possible mechanisms of MAT expansion under estrogen withdrawal and further summarize emerging findings regarding the pathological roles of MAT in bone remodeling. We also discuss the current therapies targeting MAT in osteoporosis. A comprehensive understanding of the relationship between MAT expansion and bone metabolism in estrogen deficiency conditions will provide new insights into potential therapeutic targets for PMOP.
Collapse
Affiliation(s)
- Jiao Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Chen
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lingyun Lu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China; Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
Ma B, Xu X, He S, Zhang J, Wang X, Wu P, Liu J, Jiang H, Zheng M, Li W, Wang T. STC2 modulates ERK1/2 signaling to suppress adipogenic differentiation of human bone marrow mesenchymal stem cells. Biochem Biophys Res Commun 2020; 524:163-168. [PMID: 31982135 DOI: 10.1016/j.bbrc.2020.01.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 01/10/2020] [Indexed: 01/09/2023]
Abstract
Stanniocalcin-2 (STC2) is a glycoprotein that has been found to play key roles in the regulation of cancer, diabetes mellitus, and osteogenesis. Herein we sought to extend these past studies by examining the importance of STC2 in the context of human mesenchymal stem cell (hMSC) adipogenic differentiation and exploring the mechanisms underlying such importance. We found that STC2 expression was significantly reduced on day 7 of hMSC adipogenesis. When we deliberately overexpressed STC2 in these cells, this resulted in significantly decreased expression of both peroxisome proliferator-activated receptor γ (PPARγ) and Fatty Acid Binding Protein-4 (FABP4) together with increased extracellular-signal regulated kinase 1/2 (ERK1/2) phosphorylation and markedly reduced lipid droplet formation within cells. Treatment of cells using the ERK inhibitor U0126 disrupted this ERK1/2 phosphorylation and restored the adipogenic differentiation of these hMSCs. When we instead knocked down STC2 expression, the opposite phenotypes were observed. Together these findings thus reveal that STC2 modulates ERK1/2 signaling in hMSCs so as to suppress their adipogenic differentiation.
Collapse
Affiliation(s)
- Baicheng Ma
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Xiaoyuan Xu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Shan He
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Jie Zhang
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Xinping Wang
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Ping Wu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Jianyun Liu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - He Jiang
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Meirong Zheng
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Weidong Li
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China.
| | - Tao Wang
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China.
| |
Collapse
|
29
|
Tratwal J, Labella R, Bravenboer N, Kerckhofs G, Douni E, Scheller EL, Badr S, Karampinos DC, Beck-Cormier S, Palmisano B, Poloni A, Moreno-Aliaga MJ, Fretz J, Rodeheffer MS, Boroumand P, Rosen CJ, Horowitz MC, van der Eerden BCJ, Veldhuis-Vlug AG, Naveiras O. Reporting Guidelines, Review of Methodological Standards, and Challenges Toward Harmonization in Bone Marrow Adiposity Research. Report of the Methodologies Working Group of the International Bone Marrow Adiposity Society. Front Endocrinol (Lausanne) 2020; 11:65. [PMID: 32180758 PMCID: PMC7059536 DOI: 10.3389/fendo.2020.00065] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
The interest in bone marrow adiposity (BMA) has increased over the last decade due to its association with, and potential role, in a range of diseases (osteoporosis, diabetes, anorexia, cancer) as well as treatments (corticosteroid, radiation, chemotherapy, thiazolidinediones). However, to advance the field of BMA research, standardization of methods is desirable to increase comparability of study outcomes and foster collaboration. Therefore, at the 2017 annual BMA meeting, the International Bone Marrow Adiposity Society (BMAS) founded a working group to evaluate methodologies in BMA research. All BMAS members could volunteer to participate. The working group members, who are all active preclinical or clinical BMA researchers, searched the literature for articles investigating BMA and discussed the results during personal and telephone conferences. According to the consensus opinion, both based on the review of the literature and on expert opinion, we describe existing methodologies and discuss the challenges and future directions for (1) histomorphometry of bone marrow adipocytes, (2) ex vivo BMA imaging, (3) in vivo BMA imaging, (4) cell isolation, culture, differentiation and in vitro modulation of primary bone marrow adipocytes and bone marrow stromal cell precursors, (5) lineage tracing and in vivo BMA modulation, and (6) BMA biobanking. We identify as accepted standards in BMA research: manual histomorphometry and osmium tetroxide 3D contrast-enhanced μCT for ex vivo quantification, specific MRI sequences (WFI and H-MRS) for in vivo studies, and RT-qPCR with a minimal four gene panel or lipid-based assays for in vitro quantification of bone marrow adipogenesis. Emerging techniques are described which may soon come to complement or substitute these gold standards. Known confounding factors and minimal reporting standards are presented, and their use is encouraged to facilitate comparison across studies. In conclusion, specific BMA methodologies have been developed. However, important challenges remain. In particular, we advocate for the harmonization of methodologies, the precise reporting of known confounding factors, and the identification of methods to modulate BMA independently from other tissues. Wider use of existing animal models with impaired BMA production (e.g., Pfrt-/-, KitW/W-v) and development of specific BMA deletion models would be highly desirable for this purpose.
Collapse
Affiliation(s)
- Josefine Tratwal
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Rossella Labella
- Tissue and Tumour Microenvironments Lab, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Movement Sciences, Amsterdam, Netherlands
- Section of Endocrinology, Department of Internal Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
| | - Greet Kerckhofs
- Biomechanics Lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Department Materials Engineering, KU Leuven, Leuven, Belgium
| | - Eleni Douni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, MO, United States
| | - Sammy Badr
- Univ. Lille, EA 4490 - PMOI - Physiopathologie des Maladies Osseuses Inflammatoires, Lille, France
- CHU Lille, Service de Radiologie et Imagerie Musculosquelettique, Lille, France
| | - Dimitrios C. Karampinos
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Sarah Beck-Cormier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
| | - Biagio Palmisano
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, United States
| | - Antonella Poloni
- Hematology, Department of Clinic and Molecular Science, Università Politecnica Marche-AOU Ospedali Riuniti, Ancona, Italy
| | - Maria J. Moreno-Aliaga
- Centre for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Centre of Biomedical Research Network, ISCIII, Madrid, Spain
| | - Jackie Fretz
- Department of Orthopaedics and Rehabilitation, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, CT, United States
| | - Matthew S. Rodeheffer
- Department of Comparative Medicine and Molecular, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, CT, United States
| | - Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Clifford J. Rosen
- Maine Medical Center Research Institute, Center for Clinical and Translational Research, Scarborough, ME, United States
| | - Mark C. Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Bram C. J. van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Annegreet G. Veldhuis-Vlug
- Section of Endocrinology, Department of Internal Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
- Maine Medical Center Research Institute, Center for Clinical and Translational Research, Scarborough, ME, United States
- Jan van Goyen Medical Center/OLVG Hospital, Department of Internal Medicine, Amsterdam, Netherlands
- *Correspondence: Annegreet G. Veldhuis-Vlug
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Hematology Service, Departments of Oncology and Laboratory Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Olaia Naveiras ;
| |
Collapse
|
30
|
Abstract
The skeleton harbors an array of lineage cells that have an essential role in whole body homeostasis. Adipocytes start the colonization of marrow space early in postnatal life, expanding progressively and influencing other components of the bone marrow through paracrine signaling. In this unique, closed, and hypoxic environment close to the endosteal surface and adjacent to the microvascular space the marrow adipocyte can store or provide energy, secrete adipokines, and target neighboring bone cells. Adipocyte progenitors can also migrate from the bone marrow to populate white adipose tissue, a process that accelerates during weight gain. The marrow adipocyte also has an endocrine role in whole body homeostasis through its varied secretome that targets distant adipose depots, skeletal muscle, and the nervous system. Further insights into the biology of this unique and versatile cell will undoubtedly lead to novel therapeutic approaches to metabolic and age-related disorders such as osteoporosis and diabetes mellitus.
Collapse
Affiliation(s)
- Francisco J A de Paula
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil;
| | - Clifford J Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, Maine 04074, USA;
| |
Collapse
|
31
|
Sebo ZL, Rendina-Ruedy E, Ables GP, Lindskog DM, Rodeheffer MS, Fazeli PK, Horowitz MC. Bone Marrow Adiposity: Basic and Clinical Implications. Endocr Rev 2019; 40:1187-1206. [PMID: 31127816 PMCID: PMC6686755 DOI: 10.1210/er.2018-00138] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
The presence of adipocytes in mammalian bone marrow (BM) has been recognized histologically for decades, yet, until recently, these cells have received little attention from the research community. Advancements in mouse transgenics and imaging methods, particularly in the last 10 years, have permitted more detailed examinations of marrow adipocytes than ever before and yielded data that show these cells are critical regulators of the BM microenvironment and whole-body metabolism. Indeed, marrow adipocytes are anatomically and functionally separate from brown, beige, and classic white adipocytes. Thus, areas of BM space populated by adipocytes can be considered distinct fat depots and are collectively referred to as marrow adipose tissue (MAT) in this review. In the proceeding text, we focus on the developmental origin and physiologic functions of MAT. We also discuss the signals that cause the accumulation and loss of marrow adipocytes and the ability of these cells to regulate other cell lineages in the BM. Last, we consider roles for MAT in human physiology and disease.
Collapse
Affiliation(s)
- Zachary L Sebo
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut.,Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut
| | | | - Gene P Ables
- Orentreich Foundation for the Advancement of Science, Cold Spring, New York
| | - Dieter M Lindskog
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| | - Matthew S Rodeheffer
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut.,Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut
| | - Pouneh K Fazeli
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
32
|
Hashimoto R, Miyamoto Y, Itoh S, Daida H, Okada T, Katoh Y. Phorbol 12-myristate 13-acetate (PMA) suppresses high Ca 2+-enhanced adipogenesis in bone marrow stromal cells. J Physiol Sci 2019; 69:741-748. [PMID: 31256364 PMCID: PMC10717529 DOI: 10.1007/s12576-019-00690-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022]
Abstract
We have previously reported that increased extracellular and intracellular Ca2+ lead to adipocyte accumulation in bone marrow stromal cells (BMSCs). However, strategies to suppress high Ca2+-enhanced adipocyte accumulation have not been reported. We examined the effects of the diacylglycerol analog phorbol 12-myristate 13-acetate (PMA) on proliferation and adipogenesis of mouse primary BMSCs. We used 9 mM CaCl2 and 100 nM ionomycin to increase extracellular Ca2+ and intracellular Ca2+, respectively. PMA suppressed the expression of both C/EBPα and PPARγ under normal adipogenesis, adipogenesis + CaCl2, and adipogenesis + ionomycin conditions. PMA enhanced proliferation under normal adipogenesis conditions but suppressed proliferation under adipogenesis + CaCl2 and adipogenesis + ionomycin conditions. PMA did not affect the accumulation of adipocytes under normal adipogenesis conditions but suppressed adipocyte accumulation under adipogenesis + CaCl2 and adipogenesis + ionomycin conditions. These results suggest that the PMA-dependent pathway is an important signaling pathway to suppress high Ca2+-enhanced adipocyte accumulation.
Collapse
Affiliation(s)
- Ryota Hashimoto
- Department of Physiology, Juntendo University Faculty of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Yuki Miyamoto
- Department of Cardiology, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Seigo Itoh
- Department of Cardiology, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroyuki Daida
- Department of Cardiology, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takao Okada
- Department of Physiology, Juntendo University Faculty of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Youichi Katoh
- Department of Cardiology, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Juntendo University Faculty of International Liberal Arts, Hongo 2-1-1, Bunkyo-ku, Tokyo, 112-8421, Japan.
| |
Collapse
|
33
|
Costa S, Fairfield H, Reagan MR. Inverse correlation between trabecular bone volume and bone marrow adipose tissue in rats treated with osteoanabolic agents. Bone 2019; 123:211-223. [PMID: 30954729 PMCID: PMC6559822 DOI: 10.1016/j.bone.2019.03.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 12/28/2022]
Abstract
There is currently an unmet clinical need for improved treatments for skeletal diseases such as osteoporosis and cancer-induced bone disease. This is due in part to a paucity of novel targets and an incomplete understanding of the mechanisms of action for established therapies. We defined the effects of anabolic treatments on bone and the bone marrow adipocyte (BMA). Sclerostin-neutralizing antibodies (Scl-Ab), romosozumab, human parathyroid hormone (hPTH, 1-34), and hPTH/hPTHrP analogues (e.g. teriparatide and abaloparatide) stimulate bone formation and have been studied in clinical trials for severe osteoporosis. In this study, eight-week-old male and female rats were administered vehicle, Scl-Ab (3 mg/kg or 50 mg/kg) weekly, or hPTH (1-34) (75 μg/kg) daily for 4 or 26 weeks. Histological analyses of distal femura were performed using a novel ImageJ method for trabecular bone and bone marrow adipose tissue (BMAT). Adipocyte number, circumference, and total adipose area were compared within the tissue area (T.Ar) or the marrow area (Ma.Ar), (defined as the T.Ar minus the trabecular bone area). After 26 weeks of treatment, a significant inverse correlation between bone and tissue adiposity (total adipocyte area divided by T.Ar) were observed in males and females (p < 0.0001). However, there were no significant correlations between bone and marrow adiposity (total adipocyte area divided by Ma.Ar) for either sex after 26 weeks of treatments. Scl-Ab treatments also resulted in no effect on adipocytes based on marrow adiposity for either sex after 26 weeks. However, chronic hPTH treatments significantly reduced adipocyte number and adiposity within the T.Ar and within the Ma.Ar in males. Overall, our data suggest that with long-term treatment, Scl-Abs decrease total tissue adiposity mainly by increasing trabecular bone, resulting in an overall reduction in the space in which adipocytes can reside. These findings were determined by developing and comparing two different methods of assessment of the marrow cavity, defined to either include or exclude trabecular bone. Thus, researchers should consider which adiposity measurement is more informative and relevant for their studies. Overall, our findings should help design improved therapies or combination treatments to target a potential new contributor to bone diseases: the bone marrow adipocyte.
Collapse
Affiliation(s)
- Samantha Costa
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Heather Fairfield
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Michaela R Reagan
- Maine Medical Center Research Institute, Scarborough, ME, USA; University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA; Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
34
|
Beekman KM, Veldhuis-Vlug AG, van der Veen A, den Heijer M, Maas M, Kerckhofs G, Parac-Vogt TN, Bisschop PH, Bravenboer N. The effect of PPARγ inhibition on bone marrow adipose tissue and bone in C3H/HeJ mice. Am J Physiol Endocrinol Metab 2019; 316:E96-E105. [PMID: 30457914 DOI: 10.1152/ajpendo.00265.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bone marrow adipose tissue (BMAT) increases after menopause, and increased BMAT is associated with osteoporosis and prevalent vertebral fractures. Peroxisome proliferator-activated receptor-γ (PPARγ) activation promotes adipogenesis and inhibits osteoblastogenesis; therefore, PPARγ is a potential contributor to the postmenopausal increase in BMAT and decrease in bone mass. The aim of this study is to determine if PPARγ inhibition can prevent ovariectomy-induced BMAT increase and bone loss in C3H/HeJ mice. Fourteen-week-old female C3H/HeJ mice ( n = 40) were allocated to four intervention groups: sham surgery (Sham) or ovariectomy (OVX; isoflurane anesthesia) with either vehicle (Veh) or PPARγ antagonist administration (GW9662; 1 mg·kg-1·day-1, daily intraperitoneal injections) for 3 wk. We measured BMAT volume, adipocyte size, adipocyte number. and bone structural parameters in the proximal metaphysis of the tibia using polyoxometalate-based contrast enhanced-nanocomputed topogaphy. Bone turnover was measured in the contralateral tibia using histomorphometry. The effects of surgery and treatment were analyzed by two-way ANOVA. OVX increased the BMAT volume fraction (Sham + Veh: 2.9 ± 2.7% vs. OVX + Veh: 8.1 ± 5.0%: P < 0.001), average adipocyte diameter (Sham + Veh: 19.3 ± 2.6 μm vs. OVX + Veh: 23.1 ± 3.4 μm: P = 0.001), and adipocyte number (Sham + Veh: 584 ± 337cells/μm3 vs. OVX + Veh: 824 ± 113cells/μm3: P = 0.03), while OVX decreased bone volume fraction (Sham + Veh: 15.5 ± 2.8% vs. OVX + Veh: 7.7 ± 1.9%; P < 0.001). GW9662 had no effect on BMAT, bone structural parameters, or bone turnover. In conclusion, ovariectomy increased BMAT and decreased bone volume in C3H/HeJ mice. The PPARγ antagonist GW9662 had no effect on BMAT or bone volume in C3H/HeJ mice, suggesting that BMAT accumulation is regulated independently of PPARγ in C3H/HeJ mice.
Collapse
Affiliation(s)
- Kerensa M Beekman
- Amsterdam Movement Sciences, Section of Endocrinology, Department of Internal Medicine, Vrije Universiteit, Amsterdam University Medical Center , Amsterdam , The Netherlands
- Amsterdam Movement Sciences, Department of Radiology and Nuclear Medicine, University of Amsterdam, Amsterdam University Medical Center , Amsterdam , The Netherlands
| | - Annegreet G Veldhuis-Vlug
- Amsterdam Movement Sciences, Department of Endocrinology and Metabolism, University of Amsterdam, Amsterdam University Medical Center , Amsterdam , The Netherlands
| | - Albert van der Veen
- Department of Physics and Medical Technology, Vrije Universiteit, Amsterdam University Medical Center , Amsterdam , The Netherlands
- Department Cardiology, Vrije Universiteit, Amsterdam University Medical Center , Amsterdam , The Netherlands
| | - Martin den Heijer
- Amsterdam Movement Sciences, Section of Endocrinology, Department of Internal Medicine, Vrije Universiteit, Amsterdam University Medical Center , Amsterdam , The Netherlands
| | - Mario Maas
- Amsterdam Movement Sciences, Department of Radiology and Nuclear Medicine, University of Amsterdam, Amsterdam University Medical Center , Amsterdam , The Netherlands
| | - Greet Kerckhofs
- Biomechanics Laboratory, Institute of Mechanics, Materials, and Civil Engineering, Université Catholique de Louvain , Louvain-la-Neuve , Belgium
- Department Materials Engineering, KU Leuven , Leuven , Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven , Leuven , Belgium
| | - Tatjana N Parac-Vogt
- Laboratory of Bioinorganic Chemistry, Chemistry Department, KU Leuven , Leuven , Belgium
| | - Peter H Bisschop
- Amsterdam Movement Sciences, Department of Endocrinology and Metabolism, University of Amsterdam, Amsterdam University Medical Center , Amsterdam , The Netherlands
| | - Nathalie Bravenboer
- Amsterdam Movement Sciences, Research Laboratory Bone and Calcium Metabolism, Department of Clinical Chemistry, Vrije Universiteit, Amsterdam University Medical Center , Amsterdam , The Netherlands
- Department of Internal Medicine, Leiden University Medical Center , Leiden, The Netherlands
| |
Collapse
|
35
|
Pandey MK, Gupta SC, Karelia D, Gilhooley PJ, Shakibaei M, Aggarwal BB. Dietary nutraceuticals as backbone for bone health. Biotechnol Adv 2018; 36:1633-1648. [PMID: 29597029 DOI: 10.1016/j.biotechadv.2018.03.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022]
|
36
|
Raffaele M, Barbagallo I, Licari M, Carota G, Sferrazzo G, Spampinato M, Sorrenti V, Vanella L. N-Acetylcysteine (NAC) Ameliorates Lipid-Related Metabolic Dysfunction in Bone Marrow Stromal Cells-Derived Adipocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:5310961. [PMID: 30416532 PMCID: PMC6207898 DOI: 10.1155/2018/5310961] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/27/2018] [Indexed: 12/24/2022]
Abstract
Recent experimental data suggest that fatty acids and lipotoxicity could play a role in the initiation and evolution of metabolic bone diseases such as osteoporosis. A functional bone marrow adipose tissue (BMAT) may provide support to surrounding cells and tissues or may serve as a lipid reservoir that protects skeletal osteoblasts from lipotoxicity. The present study examined the effect of N-acetylcysteine (NAC), a powerful antioxidant and precursor of glutathione, commonly used to treat chronic obstructive pulmonary disease, on triglycerides accumulation in bone marrow stromal cells-derived adipocytes. Quantification of Oil Red O stained cells showed that lipid droplets decreased following NAC treatment. Additionally, exposure of bone marrow stromal cells (HS-5) to NAC increased adiponectin, PPARγ, HO-1, and SIRT-1 and increased beta-oxidation markers such as PPARα and PPARδ mRNA levels. As there is now substantial interest in alternative medicine, the observed therapeutic value of NAC should be taken into consideration in diabetic patients.
Collapse
Affiliation(s)
- Marco Raffaele
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Ignazio Barbagallo
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Maria Licari
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Carota
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Sferrazzo
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Mariarita Spampinato
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Valeria Sorrenti
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Luca Vanella
- Department of Drug Science, Biochemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|