1
|
Wang Y, Yu X, Sun F, Fu Y, Hu T, Shi Q, Man Q. METTL14 Mediates Glut3 m6A methylation to improve osteogenesis under oxidative stress condition. Redox Rep 2025; 30:2435241. [PMID: 39737912 DOI: 10.1080/13510002.2024.2435241] [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] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVES Bone remodeling imbalance contributes to osteoporosis. Though current medications enhance osteoblast involvement in bone formation, the underlying pathways remain unclear. This study was aimed to explore the pathways involved in bone formation by osteoblasts, we investigate the protective role of glycolysis and N6-methyladenosine methylation (m6A) against oxidative stress-induced impairment of osteogenesis in MC3T3-E1 cells. METHODS We utilized a concentration of 200 μM hydrogen peroxide (H2O2) to establish an oxidative damage model of MC3T3-E1 cells. Subsequently, we examined the alterations in the m6A methyltransferases (METTL3, METTL14), glucose transporter proteins (GLUT1, GLUT3) and validated m6A methyltransferase overexpression in vitro and in an osteoporosis model. The osteoblast differentiation and osteogenesis-related molecules and serum bone resorption markers were measured by biochemical analysis, Alizarin Red S staining, Western blot and ELISA. RESULTS H2O2 treatment inhibited glycolysis and osteoblast differentiation in MC3T3-E1 cells. However, when METTL14 was overexpressed, these changes induced by H2O2 could be mitigated. Our findings indicate that METTL14 promotes GLUT3 expression via YTHDF1, leading to the modulation of various parameters in the H2O2-induced model. Similar positive effects of METTL14 on osteogenesis were observed in an ovariectomized mouse osteoporosis model. DISCUSSION METTL14 could serve as a potential therapeutic approach for enhancing osteoporosis treatment.
Collapse
Affiliation(s)
- Ying Wang
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xueying Yu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Fenyong Sun
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yan Fu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Tingting Hu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qiqing Shi
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qiuhong Man
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Tint MT, Cremaschi A, Leow MKS, Padmapriya N, Ang SB, Lai JS, Chan JKY, Bernard JY, Gluckman PD, Chong YS, Godfrey KM, Müller-Riemenschneider F, Zhang C, Harvey NC, De Iorio M, Eriksson JG. Differential contributions of lean and fat mass on bone mineral density in Asian women of reproductive age: the Singapore Preconception Study of Long-Term Maternal and Child Outcomes study. JBMR Plus 2025; 9:ziaf054. [PMID: 40390807 PMCID: PMC12087953 DOI: 10.1093/jbmrpl/ziaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 05/21/2025] Open
Abstract
The relationships between fat mass (FM), lean mass (LM), and bone mass are complex with significant implications for obesity, sarcopenia, and osteoporosis later in life. While greater LM is associated with higher BMD, the association between FM and BMD is less clear. Such relationships warrant further investigation, especially in Asians, who have a higher risk of metabolic diseases and osteoporotic fractures compared to Western populations. This study investigated the associations of LM, FM, and modifiable risk factors with BMD in Asian women aged 18-45 yr. A total of 191 women from the Singapore Preconception Study of Long-Term Maternal and Child Outcomes (S-PRESTO) cohort study underwent DXA scanning at the first study visit for BMD and body composition measurements. LM, FM, and four body composition phenotypes derived from dichotomizing LM and FM were related to cohort-specific Z-scores of BMD at FN (BMDFN), LS (BMDLS), and whole body (BMDWB). Adjusting for covariates, LM showed positive associations with Z-BMDFN, [β (95%CI)], [0.38 (0.22, 0.55)], Z-BMDLS [0.43 (0.25, 0.62)], and Z-BMDWB, [0.63 (0.44, 0.81)]. Fat mass by contrast showed an inverse association only with Z-BMDWB, [-0.39 (-0.58, -0.20)]. Compared to women with healthy body composition (higher LM-lower FM), women with lower LM-higher FM had similar BMI, mean (SD) 20.9 (1.5) kg/m2 but disproportionately higher percent fat, 38.4 (2.2%), and lower Z-BMDFN [-0.58 (-0.97, -0.18)], Z-BMDLS [-0.41 (-0.81, 0.00)], and Z-BMDWB [-0.66 (-1.06, -0.25)]. Chinese women had lower BMD than Malay women. Physical activity and education attainment were positively, while the age of menarche was negatively associated with BMD. These findings in young women underscore the importance of early interventions recognizing ethnic differences in BMD to improve lifecourse musculoskeletal health. Most importantly, intervention strategies for Asian women should focus on healthy body composition beyond BMI, with a goal to preserve or increase LM.
Collapse
Affiliation(s)
- Mya Thway Tint
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (ASTAR), Singapore 117609, Republic of Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Andrea Cremaschi
- School of Science and Technology, IE University, 28006 Madrid, Spain
| | - Melvin Khee Shing Leow
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (ASTAR), Singapore 117609, Republic of Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore
- Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (ASTAR), Singapore 138669, Republic of Singapore
- Metabolic Disorders Research Programme, Lee Kong Chian School of Medicine, Singapore 308232, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Natarajan Padmapriya
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Seng Bin Ang
- Family Medicine Service, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
| | - Jun Shi Lai
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (ASTAR), Singapore 117609, Republic of Singapore
| | - Jerry Kok Yen Chan
- Department of Obstetrics and Gynaecology, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
| | - Jonathan Y Bernard
- Centre for Research in Epidemiology and StatisticS (CRESS), Université Paris Cité, Université Paris Centre Nord, Inserm, INRAE, F-75004 Paris, France
| | - Peter D Gluckman
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (ASTAR), Singapore 117609, Republic of Singapore
- Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Yap-Seng Chong
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (ASTAR), Singapore 117609, Republic of Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, NHS Foundation Trust, Southampton SO16 6YD, United Kingdom
| | - Falk Müller-Riemenschneider
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
- Berlin Institute of Health, Charite University Medical Centre, 10117 Berlin, Germany
| | - Cuilin Zhang
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Global Center for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, NHS Foundation Trust, Southampton SO16 6YD, United Kingdom
| | - Maria De Iorio
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (ASTAR), Singapore 117609, Republic of Singapore
- Department of Paediatrics, National University of Singapore, Singapore 119228, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (ASTAR), Singapore 117609, Republic of Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, PO Box 20, 00290 Helsinki, Finland
- Folkhälsan Research Center, 00290 Helsinki, Finland
| |
Collapse
|
3
|
Jafariandehkordi A, Jafariandehkordi Z. A biomechanical assessment of dental implant stability under the effect of changes in bone remodeling factors and implant elastic modulus. Comput Biol Med 2025; 192:110318. [PMID: 40367627 DOI: 10.1016/j.compbiomed.2025.110318] [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: 09/20/2024] [Revised: 03/20/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND AND OBJECTIVE The simulation of the biological phenomena can assist physicians in development of the advanced therapeutic plans and methods based on the computer model results and engineering analysis. The application of these models can predict and evaluate the medical experiment output by removing the barriers of technical and practical limitations, and expensive laboratory equipment. This provides the scholars with the preview and insight to design practical experiments and save cost, energy, and time. The present study aimed to provide a calculational, finite element-based simulation of the mandible bone's remodeling under the osteoporosis condition to quantify and evaluate the stability of a dental implant using the indicator implant stability quotient (ISQ). The effects of the significant chemical factors involved in bone remodeling including osteoprotegerin (OPG), transforming growth factor beta (TGF-β), and the parathyroid hormone (PTH) as well as the effect of the implant's elastic modulus were also included in the model. The literature lacks a predictive combinatory model considering the comparative effects of these chemicals on the implant stability which was addressed by this study. METHODS A 3D geometry of the mandible portion assembled with a Straumann implant geometry was developed using a CT image dataset. The model geometries were imported to finite element software for the definition of the material properties, boundary conditions, constraints, and mathematical relations. The model's basic parameters were based on other published works. A mesh of the geometry was created and bone remodeling partial differential equations were incorporated. The mastication force was assumed constant and the damping factor was considered zero in our study. Finally, the calculated resonance frequencies of the implant-mandible portion assembly were utilized to obtain and assess the implant stability quotient. RESULTS The results showed that a 6 % increase in the dosage rates of TGF-β had the highest increase in ISQ values to 24.28 % from the baseline and the implant elastic modulus of 12.5 GPa caused a 21.55 % relative growth in the ISQ value of the baseline. The accuracy of the results was tested by the mesh convergence and sensitivity studies. CONCLUSIONS Based on the results, TGF-β had the most significant effect on the growth of ISQ, and the implant elastic modulus remarkably increased ISQ values when its value was close to the average values of the mandible equivalent elastic modulus.
Collapse
|
4
|
Samanta A, Yoo MJ, Koh J, Lufkin SC, Lufkin T, Kraus P. Proteomic profiling of small extracellular vesicles from bovine nucleus pulposus cells. PLoS One 2025; 20:e0324179. [PMID: 40440285 PMCID: PMC12121814 DOI: 10.1371/journal.pone.0324179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/21/2025] [Indexed: 06/02/2025] Open
Abstract
Small extracellular vesicles (small EV) are a conserved means of communication across the domains of life and lately gained more interest in mammalian non-cancerous work as non-cellular, biological therapeutic with encouraging results in recent studies of chronic degenerative diseases. The nucleus pulposus (NP) is the avascular and aneural center of an intervertebral disc (IVD), home to unique niche conditions and affected in IVD degeneration. We investigated autologous and mesenchymal stem cell (MSC) small EVs for their potential to contribute to cell and tissue homeostasis in the NP niche via mass spectrometric proteome and functional enrichment analysis using adult and fetal donors. We compared these findings to published small EV databases and MSC small EV data. We propose several mechanisms associated with NP small EVs: Membrane receptor trafficking to modify signal responses promoting niche homeostasis; Redox and energy homeostasis via metabolic enzymes delivery; Cell homeostasis via proteasome delivery and immunomodulation beyond an association with a serum protein corona. The proteome signature of small EVs generated by NP parent cells is similar to previously published small EV data, yet with a focus on supplementing anaerobic metabolism and redox balance while contributing to the maintenance of an aneural and avascular microniche.
Collapse
Affiliation(s)
- Ankita Samanta
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Mi-Jeong Yoo
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Jin Koh
- The Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States of America
| | - Sina Charlotte Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Thomas Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| |
Collapse
|
5
|
Ahmed F, Ahmad SS, Alam MM, Shaquiquzzaman M, Altamish M, Krishnan A, Vohora D, Najmi AK, Khan MA. Osteogenic effect of alogliptin in chemical-induced bone loss: a tri-modal in silico, in vitro, and in vivo analysis. J Pharm Pharmacol 2025; 77:668-684. [PMID: 39360980 DOI: 10.1093/jpp/rgae112] [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: 02/22/2024] [Accepted: 08/13/2024] [Indexed: 05/03/2025]
Abstract
OBJECTIVE To investigate the effects of Alogliptin in chemical-induced post-menopausal osteoporosis. METHODOLOGY The binding affinity of alogliptin with osteogenic proteins was analysed in silico. The effect of alogliptin on osteogenic proteins and mineralization of osteoblastic cells was evaluated in UMR-106 cells. Further, in vivo anti-osteoporotic activity of alogliptin was evaluated in postmenopausal osteoporosis. Various bone turnover markers were assayed in serum. This followed the analysis of microarchitecture of bone, histology, and immunohistochemistry (IHC) of bone tissue. RESULTS Docking scores showed that alogliptin has binding affinity for bone alkaline phosphatase (BALP), osteocalcin, and bone morphogenic protein (BMP-2). Alogliptin also enhanced mineralization of osteoblast cells, evidenced with increased ALP, osteocalcin, and BMP-2. Animal studies revealed significant elevation of bone formation markers, bone ALP, osteocalcin and BMP-2, and decreased bone resorption markers, receptor activator of NF-κβ (RANKL), cathepsin K (CTSK), tartrate resistant acid phosphatase (TRAcP5b) in VCD-induced post-menopausal osteoporosis. Micro computed tomography (μCT) analysis and histology of femur bone and lumbar vertebrae demonstrated decrease in trabecular separation and improved bone density. IHC of femur showed reduced DPP4 enzyme. CONCLUSIONS Alogliptin increased mineralization in osteoblast cells. It had beneficial effects also altered bone turnover markers, repaired the trabecular microstructure, improved bone mineral density, and exhibited bone forming capacity targeting DPP-4 enzyme in postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Faraha Ahmed
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Sufian Ahmad
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Shaquiquzzaman
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Altamish
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Anuja Krishnan
- Department of Molecular Medicine, School of Interdisciplinary Science and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
6
|
Dayal A, Gray M, Vallejo JA, Lara-Castillo N, Johnson ML, Wacker MJ. MLO-Y4 fluid flow shear stress conditioned media enhances cardiac contractility and intracellular Ca 2. Am J Physiol Regul Integr Comp Physiol 2025; 328:R591-R600. [PMID: 40135808 DOI: 10.1152/ajpregu.00287.2024] [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: 11/21/2024] [Revised: 01/05/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
The skeleton is in complex interplay with the other systems of the body and is highly responsive to input from the external environment. Bone mechanical loading results in interstitial fluid flow via the lacunar-canalicular system, generating fluid flow sheer stress (FFSS). FFSS variably stresses osteocytes, subsequently causing the release of metabolites and protein factors that function locally to increase bone formation and may play a role in cross talk between various organ systems, for instance between bone and skeletal muscle. Therefore, we hypothesized that this cross talk includes altering cardiac function. To test this hypothesis, media conditioned by MLO-Y4 osteocyte-like cell culture line under FFSS was used to model the endocrine effects of bone during mechanical loading on contraction of ex vivo Langendorf-perfused isolated hearts. When hearts were externally paced at a fixed rate, FFSS osteocyte conditioned media (CM) induced significant premature contractions compared with vehicle (control). FFSS osteocyte CM administration to self-paced hearts increased total contraction force by 31%. To determine whether the mechanism involved intracellular Ca2+, vehicle and FFSS bone CM were perfused over cultured H9C2 cardiomyocytes while undergoing Ca2+ imaging using Fluo-8. We observed an increase in intracellular Ca2+ with FFSS CM perfusion of cardiomyocytes compared with vehicle. These increases were only present with exogenous electrical pacing. Our findings demonstrate that FFSS bone CM enhances cardiac contractility by increasing intracellular cardiomyocyte Ca2+. The results obtained in this study suggest that the skeleton, responding to mechanical strain, has the potential to augment cardiac output and provide evidence for bone-heart cross talk.NEW & NOTEWORTHY The skeletal system operates as an endocrine organ, releasing factors that impact multi-tissue physiology. The results obtained in this study demonstrate that conditioned media collected from MLO-Y4 osteocytes exposed to fluid flow shear stress increases cardiomyocyte intracellular calcium and enhances cardiac contractility in vitro. These results support the concept of bone-heart cross talk that may have implications in exercise training, reduced-function settings such as bedrest, and the interplay between bone and heart health.
Collapse
Affiliation(s)
- Anuhya Dayal
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, United States
| | - Mark Gray
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, United States
| | - Julian A Vallejo
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, United States
- Department of Oral & Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, United States
| | - Nuria Lara-Castillo
- Department of Oral & Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, United States
| | - Mark L Johnson
- Department of Oral & Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, United States
| | - Michael J Wacker
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, United States
| |
Collapse
|
7
|
Henry ÓC, O'Neill LAJ. Metabolic Reprogramming in Stromal and Immune Cells in Rheumatoid Arthritis and Osteoarthritis: Therapeutic Possibilities. Eur J Immunol 2025; 55:e202451381. [PMID: 40170391 PMCID: PMC11962241 DOI: 10.1002/eji.202451381] [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: 12/03/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/03/2025]
Abstract
Metabolic reprogramming of stromal cells, including fibroblast-like synoviocytes (FLS) and chondrocytes, as well as osteoclasts (OCs), are involved in the inflammatory and degenerative processes underlying rheumatoid arthritis (RA) and osteoarthritis (OA). In RA, FLS exhibit mTOR activation, enhanced glycolysis and reduced oxidative phosphorylation, fuelling inflammation, angiogenesis, and cartilage degradation. In OA, chondrocytes undergo metabolic rewiring, characterised by mTOR and NF-κB activation, mitochondrial dysfunction, and increased glycolysis, which promotes matrix metalloproteinase production, extracellular matrix (ECM) degradation, and angiogenesis. Macrophage-derived immunometabolites, including succinate and itaconate further modulate stromal cell function, acting as signalling molecules that modulate inflammatory and catabolic processes. Succinate promotes inflammation whilst itaconate is anti-inflammatory, suppressing inflammatory joint disease in models. Itaconate deficiency also correlates inversely with disease severity in RA in humans. Emerging evidence highlights the potential of targeting metabolic processes as promising therapeutic strategies for connective tissue disorders.
Collapse
Affiliation(s)
- Órlaith C. Henry
- Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | | |
Collapse
|
8
|
Abu-Nada L, Liu Y, Saleh Al-Hamed F, Ouliass B, Millecamps M, Tran SD, Ferland G, Soleimani VD, Marino FT, Murshed M. Young bone marrow transplantation delays bone aging in old mice. Exp Gerontol 2025; 202:112704. [PMID: 39914580 DOI: 10.1016/j.exger.2025.112704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Recent discoveries have shown that systemic manipulations, such as parabiosis, blood exchange, and young plasma transfer, can counteract many hallmarks of aging. This rejuvenation effect has been attributed to circulatory factors produced by cells from both hematopoietic and non-hematopoietic lineages. However, the specific involvement of bone marrow (BM) or hematopoietic cells in producing such factors and their effects on aging is still unclear. We developed a model of aged mice with transplanted young or old BM cells and assessed the impact on the aging process, specifically on energy metabolism and bone remodeling parameters. The donor BM cell engraftment in the aged mice was confirmed by flow cytometry using a transplanted cell-specific marker (green fluorescent protein). Energy metabolism was assessed using Oxymax indirect calorimetry system after 3 months of transplantation. Tibiae and L3-L4 vertebrae were analyzed using micro-CT, a three-point bending test and bone histomorphometry. Moreover, bone marrow proteome was assessed using proteomics, and blood serum/plasma was collected and analyzed using the Luminex assay. Our results showed that while the effect on energy metabolism was insignificant, rejuvenating the BM through young bone marrow transplantation reversed age-associated low bone mass traits in old mice. Specifically, young bone marrow transplantation improved bone trabecular microarchitecture both in tibiae and vertebrae of old mice and increased the number of osteoblasts and osteoclasts compared to old bone marrow transplantation. In conclusion, young bone marrow cells may represent a future therapeutic strategy for age-related diseases such as osteoporosis. The findings of this study provide important insights into our understanding of aging.
Collapse
Affiliation(s)
- Lina Abu-Nada
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Younan Liu
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | | | - Bouchra Ouliass
- Montreal Heart Institute Research Centre, Montreal, QC, Canada
| | - Magali Millecamps
- ABC-Platform (Animal Behavioral Characterization) at Alan Edwards Center for Research on Pain, McGill University, Montreal, Quebec, Canada; Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | | | - Vahab D Soleimani
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | | | - Monzur Murshed
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Shriners hospital for children, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Kang H, Peng R, Dong Y, Liao F, Zhu M, Wang P, Hu SA, Hu P, Wang J, Liu Z, Song K, Li F. TRAF1 promotes osteoclastogenesis by enhancing metabolic adaptation to oxidative phosphorylation in an AKT-dependent manner. Mol Ther 2025; 33:933-949. [PMID: 39863932 PMCID: PMC11897774 DOI: 10.1016/j.ymthe.2025.01.040] [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: 09/06/2024] [Revised: 12/01/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025] Open
Abstract
Tumor necrosis factor receptor-associated factor 1 (TRAF1) is a crucial signaling adaptor involved in multiple cellular events. However, its role in regulating osteoclastogenesis and energy metabolism remains unclear. Here, we report that TRAF1 promotes osteoclastogenesis and oxidative phosphorylation (OXPHOS). Employing RNA sequencing, we found that TRAF1 is markedly upregulated during osteoclastogenesis and is positively associated with osteoporosis. TRAF1 knockout inhibits osteoclastogenesis and increases bone mass in both normal and ovariectomized adult mice without affecting bone mass in childhood. Furthermore, TRAF1 promotes osteoclast OXPHOS by increasing the phosphorylation level of AKT. Mechanistically, TRAF1 functions to inhibit TRAF2-induced ubiquitination of Gβl, a known activator of AKT, and further upregulates AKT phosphorylation. Rescue experiments revealed that the inhibitory effects of TRAF1 knockout on osteoclastogenesis, OXPHOS, and bone mass are dependent on AKT. Collectively, our findings uncover a previously unrecognized function of TRAF1 in regulating osteoclastogenesis and energy metabolism, and establish a novel TRAF1-AKT-OXPHOS axis in osteoclasts.
Collapse
Affiliation(s)
- Honglei Kang
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Research Institute of Shenzhen Huazhong University of Science and Technology, Shen Zhen, China
| | - Renpeng Peng
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yimin Dong
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuben Liao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, China
| | - Meipeng Zhu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengju Wang
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-An Hu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peixuan Hu
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Wang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zheming Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, China.
| | - Kehan Song
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, China.
| | - Feng Li
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
Liu X, Hu F, Zhang Y, Ma S, Liu H, Shang D, Yin P. Metabolomics Approach Revealed Polyunsaturated Fatty Acid Disorders as Pathogenesis for Chronic Pancreatitis-Induced Osteoporosis in Mice. Metabolites 2025; 15:173. [PMID: 40137138 PMCID: PMC11944031 DOI: 10.3390/metabo15030173] [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: 01/10/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Osteoporosis is frequently observed in patients with chronic pancreatitis, and both conditions are closely associated with systemic metabolic disorders. However, the underlying mechanisms linking chronic pancreatitis and osteoporosis remain unclear. Methods: In this study, we utilized high-performance liquid chromatography-mass spectrometry (HPLC-MS) to conduct metabolomics and lipidomics analyses on pancreatic, serum, and other tissues from a mouse model of chronic pancreatitis-induced osteoporosis (CP-OP), with the aim to elucidate the metabolism-related pathogenic mechanisms of CP-OP. Results: We identified over 405 metabolites and 445 lipids, and our findings revealed that several metabolites involving the tricarboxylic acid (TCA) cycle, as well as triacylglycerols and diacylglycerols with higher saturation, were significantly increased in the CP-OP model. In contrast, triglycerides with higher unsaturation were decreased. Differential pathways were enriched in n-3 long-chain polyunsaturated fatty acid metabolism in both pancreatic and bone tissues, and these pathways exhibited positive correlations with bone-related parameters. Furthermore, the modulation of these polyunsaturated fatty acids by Qingyi granules demonstrated significant therapeutic effects on CP-OP, as validated in mouse models. Conclusions: Through the metabolomics approach, we uncovered that disorders in polyunsaturated fatty acids play a critical role in the pathogenesis of CP-OP. This study not only enhances our understanding of the pathogenesis of CP-OP but also highlights the therapeutic potential of targeting polyunsaturated fatty acids as a future intervention strategy for osteoporosis treatment.
Collapse
Affiliation(s)
- Xinlin Liu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Fenglin Hu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yunshu Zhang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Shurong Ma
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Haihua Liu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Guantao County People’s Hospital, Handan 057750, China
| | - Dong Shang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Peiyuan Yin
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| |
Collapse
|
11
|
Jiang Y, Ye AH, He WG, Liu L, Gao X, Liu H, Liu WT, Ye FL, He DM, Liao JY, Wang J, He BC. Reducing PDK4 level constitutes a pivotal mechanism for glucocorticoids to impede osteoblastic differentiation through the enhancement of ferroptosis in mesenchymal stem cells. Stem Cell Res Ther 2025; 16:91. [PMID: 40001240 PMCID: PMC11863902 DOI: 10.1186/s13287-025-04186-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND This study mainly explores the possible role and mechanism of pyruvate dehydrogenase kinase 4 (PDK4) in the onset and development of Glucocorticoid-induced osteoporosis (GIOP), and seeks potential targets for the treatment of GIOP. METHODS Mesenchymal stem cells (MSCs) were treated with osteogenic induction medium. An in vitro osteogenic damage model was established by exposing MSCs to a high concentration (10- 6 M) of dexamethasone (DEX). Osteogenic markers were measured with real-time quantitative polymerase chain reaction, western blot, alkaline phosphatase staining, and Alizarin Red S staining. Ferroptosis markers were assessed through reactive oxygen species (ROS) fluorescent probe, transmission electron microscopy, and measurement of malondialdehyde (MDA). The potential mechanism was investigated using RT-qPCR, western blot, lysosomal probes, molecular docking, and other analytical approaches. The role of PDK4 was validated by using a GIOP rat model, micro-computed tomography and Masson's trichrome staining. RESULTS High concentrations (10- 6 M) of DEX inhibited osteogenic differentiation in C3H10T1/2 cells, and PDK4 exhibited the opposite effect. PDK4 partially reversed the osteogenic inhibitory effect of DEX both in vivo and in vitro. DEX caused mitochondrial shrinkage and disappearance of cristae in C3H10T1/2 cells, as well as an increase in total iron, ROS, MDA contents, and the level of ferroptosis key factors. These changes were partially weakened by PDK4. The ferroptosis inhibitor ferrostatin-1 partially blocked the inhibitory effect of DEX, while ferroptosis inducer RSL3 inhibited osteogenic differentiation and weakened the reversal effect of PDK4. DEX reduced the protein level of PDK4, which was partially weakened by Bafilomycin A1. The molecular docking results showed that DEX can directly bind with PDK4. CONCLUSION PDK4 can enhance the osteogenic differentiation ability of MSCs and bone mass of GIOP rats. DEX may promote the degradation of PDK4 via lysosome pathway, through which to weaken the osteogenic ability of MSCs by increasing ferroptosis. PDK4 may become a potential target for improving GIOP.
Collapse
Affiliation(s)
- Yue Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Ai-Hua Ye
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wen-Ge He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
- Department of Orthropetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lu Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiang Gao
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Department of Orthropetics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hang Liu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Department of Orthropetics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wen-Ting Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Fang-Lin Ye
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dong-Mei He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jun-Yi Liao
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Jing Wang
- Department of Blood Transfusion, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
| | - Bai-Cheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
12
|
Palomino Lago E, Ross AKC, McClellan A, Guest DJ. Identification of a global gene expression signature associated with the genetic risk of catastrophic fracture in iPSC-derived osteoblasts from Thoroughbred horses. Anim Genet 2025; 56:e13504. [PMID: 39801206 PMCID: PMC11726005 DOI: 10.1111/age.13504] [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: 11/18/2024] [Revised: 11/18/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Bone fractures are a significant problem in Thoroughbred racehorses. The risk of fracture is influenced by both genetic and environmental factors. To determine the biological processes that are affected in genetically susceptible horses, we utilised polygenic risk scoring to establish induced pluripotent stem cells (iPSCs) from horses at high and low genetic risk. RNA-sequencing on iPSC-derived osteoblasts revealed 112 genes that were significantly differentially expressed. Forty-three of these genes have known roles in bone, 27 are not yet annotated in the equine genome and 42 currently have no described role in bone. However, many of the proteins encoded by the known and unknown genes have reported interactions. Functional enrichment analyses revealed that the differentially expressed genes were overrepresented in processes regulating the extracellular matrix and pathways known to be involved in bone remodelling and bone diseases. Gene set enrichment analysis also detected numerous biological processes and pathways involved in glycolysis with the associated genes having a higher expression in the iPSC-osteoblasts from horses with low polygenic risk scores for fracture. Therefore, the differentially expressed genes may be relevant for maintaining bone homeostasis and contribute to fracture risk. A deeper understanding of the consequences of mis-regulation of these genes and the identification of the DNA variants which underpin their differential expression may reveal more about the molecular mechanisms which are involved in equine bone health and fracture risk.
Collapse
Affiliation(s)
- Esther Palomino Lago
- Department of Clinical Sciences and Services, Centre for Vaccinology and Regenerative MedicineThe Royal Veterinary CollegeHatfieldHertsUK
| | - Amy K. C. Ross
- Department of Clinical Sciences and Services, Centre for Vaccinology and Regenerative MedicineThe Royal Veterinary CollegeHatfieldHertsUK
| | - Alyce McClellan
- Animal Health TrustNewmarketUK
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - Deborah J. Guest
- Department of Clinical Sciences and Services, Centre for Vaccinology and Regenerative MedicineThe Royal Veterinary CollegeHatfieldHertsUK
| |
Collapse
|
13
|
Liu Q, Xue Y, Guo J, Tao L, Zhu Y. Citrate: a key signalling molecule and therapeutic target for bone remodeling disorder. Front Endocrinol (Lausanne) 2025; 15:1512398. [PMID: 39886032 PMCID: PMC11779597 DOI: 10.3389/fendo.2024.1512398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/20/2024] [Indexed: 02/01/2025] Open
Abstract
Bone remodeling is a continuous cyclic process that maintains and regulates bone structure and strength. The disturbance of bone remodeling leads to a series of bone metabolic diseases. Recent studies have shown that citrate, an intermediate metabolite of the tricarboxylic acid (TCA) cycle, plays an important role in bone remodeling. But the exact mechanism is still unclear. In this study, we focused on the systemic regulatory mechanism of citrate on bone remodeling, and found that citrate is involved in bone remodeling in multiple ways. The participation of citrate in oxidative phosphorylation (OXPHOS) facilitates the generation of ATP, thereby providing substantial energy for bone formation and resorption. Osteoclast-mediated bone resorption releases citrate from bone mineral salts, which is subsequently released as an energy source to activate the osteogenic differentiation of stem cells. Finally, the differentiated osteoblasts secrete into the bone matrix and participate in bone mineral salts formation. As a substrate of histone acetylation, citrate regulates the expression of genes related to bone formation and bone reabsorption. Citrate is also a key intermediate in the metabolism and synthesis of glucose, fatty acids and amino acids, which are three major nutrients in the organism. Citrate can also be used as a biomarker to monitor bone mass transformation and plays an important role in the diagnosis and therapeutic evaluation of bone remodeling disorders. Citrate imbalance due to citrate transporter could result in the supression of osteoblast/OC function through histone acetylation, thereby contributing to disorders in bone remodeling. Therefore, designing drugs targeting citrate-related proteins to regulate bone citrate content provides a new direction for the drug treatment of diseases related to bone remodeling disorders.
Collapse
Affiliation(s)
| | | | | | - Lin Tao
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Yue Zhu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Saki F, Rahimikashkooli N, Masjedi M, Dastghaib S, Koohpeyma F. Gender-Specific effects of L-arginine supplementation on bone mineral density and trabecular bone volume in Sprague-Dawley rats; stereological study. BMC Complement Med Ther 2024; 24:425. [PMID: 39725944 DOI: 10.1186/s12906-024-04736-3] [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: 11/09/2023] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND L-arginine (Arg) is a semi-essential amino acid that can be used as a key mediator for the release of growth hormone (GH), insulin-like growth factor-1(IGF-1), and other growth factors. In this study, we comprehensively evaluated the effect of Arg intake on bone growth and associated markers. METHODS The study involved 24 Sprague-Dawley rats (12 males, 12 females) divided into two groups (Age = 24 days). One group received a standard diet, while the other was injected with 10 mg/kg of Arg daily for 90 days. Serum bone markers like calcium (Ca), phosphorous(P), and alkaline phosphatase (ALP) were analyzed via colorimetric assays. stereological study and bone mineral density (BMD) were conducted via dissector method and Hologic Dual-energy x-ray absorptiometry (DXA) system; respectively. RESULTS Biochemical assays showed no significant differences in Ca, P, and ALP levels between groups. Male rats in the case group exhibited lower testosterone levels (p.value = 0.009). Stereological and bone mineral density (BMD) analyses revealed contrasting gender-specific outcomes. Female rats in the case group had higher BMD (p.value = 0.001), while males had lower BMD compared to controls (p.value = 0.018). Arg consumption affects trabecula volume values differently in females compared to males (p.value = 0.022). Furthermore, the study observed decreased osteocytes and osteoblasts in male case rats. The gender-based differences in BMD were attributed to Arg's paradoxical impact on testosterone levels in males. CONCLUSION Overall, Arg supplementation was found to influence BMD and trabecular bone volume, with outcomes varying depending on gender. The study highlights the intricate interplay between Arg, sex hormones, and bone health, offering insights into these complex relationships.
Collapse
Affiliation(s)
- Forough Saki
- Pediatric Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, P.O. Box, Shiraz, 71345-1744, Iran
| | - Nima Rahimikashkooli
- Internal Medicine Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Moein Masjedi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Dastghaib
- Pediatric Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, P.O. Box, Shiraz, 71345-1744, Iran.
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Clinical Biochemistry in Endocrine and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Farhad Koohpeyma
- Pediatric Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, P.O. Box, Shiraz, 71345-1744, Iran.
- Medical Physiology, Shiraz Endocrine and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
Jia X, Zhang G, Yu D. Application of extracellular vesicles in diabetic osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1466775. [PMID: 39720256 PMCID: PMC11666354 DOI: 10.3389/fendo.2024.1466775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/15/2024] [Indexed: 12/26/2024] Open
Abstract
As the population ages, the occurrence of osteoporosis is becoming more common. Diabetes mellitus is one of the factors in the development of osteoporosis. Compared with the general population, the incidence of osteoporosis is significantly higher in diabetic patients. Diabetic osteoporosis (DOP) is a metabolic bone disease characterized by abnormal bone tissue structure due to hyperglycemia and insulin resistance, reduced bone strength and increased risk of fractures. This is a complex mechanism that occurs at the cellular level due to factors such as blood vessels, inflammation, and hyperglycemia and insulin resistance. Although the application of some drugs in clinical practice can reduce the occurrence of DOP, the incidence of fractures caused by DOP is still very high. Extracellular vesicles (EVs) are a new communication mode between cells, which can transfer miRNAs and proteins from mother cells to target cells through membrane fusion, thereby regulating the function of target cells. In recent years, the role of EVs in the pathogenesis of DOP has been widely demonstrated. In this article, we first describe the changes in the bone microenvironment of osteoporosis. Second, we describe the pathogenesis of DOP. Finally, we summarize the research progress and challenges of EVs in DOP.
Collapse
Affiliation(s)
- Xiaopeng Jia
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Gongzi Zhang
- Department of Rehabilitation Medicine, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Deshui Yu
- Trauma Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
16
|
Bellman J, Sjöros T, Hägg D, Atencio Herre E, Hieta J, Eskola O, Laitinen K, Nuutila P, Jansson JO, Jansson PA, Kalliokoski K, Roivainen A, Ohlsson C. Loading Enhances Glucose Uptake in Muscles, Bones, and Bone Marrow of Lower Extremities in Humans. J Clin Endocrinol Metab 2024; 109:3126-3136. [PMID: 38753869 PMCID: PMC11570666 DOI: 10.1210/clinem/dgae344] [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: 03/08/2024] [Revised: 04/29/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
CONTEXT Increased standing time has been associated with improved health, but the underlying mechanism is unclear. OBJECTIVES We herein investigate if increased weight loading increases energy demand and thereby glucose uptake (GU) locally in bone and/or muscle in the lower extremities. METHODS In this single-center clinical trial with a randomized crossover design (ClinicalTrials.gov ID, NCT05443620), we enrolled 10 men with body mass index between 30 and 35 kg/m2. Participants were treated with both high load (standing with weight vest weighing 11% of body weight) and no load (sitting) on the lower extremities. GU was measured using whole-body quantitative positron emission tomography/computed tomography imaging. The primary endpoint was the change in GU ratio between loaded bones (ie, femur and tibia) and nonloaded bones (ie, humerus). RESULTS High load increased the GU ratio between lower and upper extremities in cortical diaphyseal bone (eg, femur/humerus ratio increased by 19%, P = .029), muscles (eg, m. quadriceps femoris/m. triceps brachii ratio increased by 28%, P = .014), and certain bone marrow regions (femur/humerus diaphyseal bone marrow region ratio increased by 17%, P = .041). Unexpectedly, we observed the highest GU in the bone marrow region of vertebral bodies, but its GU was not affected by high load. CONCLUSION Increased weight-bearing loading enhances GU in muscles, cortical bone, and bone marrow of the exposed lower extremities. This could be interpreted as increased local energy demand in bone and muscle caused by increased loading. The physiological importance of the increased local GU by static loading remains to be determined.
Collapse
Affiliation(s)
- Jakob Bellman
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-41390 Gothenburg, Sweden
| | - Tanja Sjöros
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
| | - Daniel Hägg
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-41390 Gothenburg, Sweden
| | - Erika Atencio Herre
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
| | - Janina Hieta
- Nutrition and Food Research Center and Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, Faculty of Medicine, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Olli Eskola
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Kirsi Laitinen
- Nutrition and Food Research Center and Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, Faculty of Medicine, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
- Department of Endocrinology, Turku University Hospital, FI-20520 Turku, Finland
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-41390 Gothenburg, Sweden
| | - Per-Anders Jansson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-41345 Gothenburg, Sweden
- Gothia Forum, Region Västra Götaland, Sahlgrenska University Hospital, SE-41346 Gothenburg, Sweden
| | - Kari Kalliokoski
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
- InFLAMES Research Flagship, University of Turku, FI-20014 Turku, Finland
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-41345 Gothenburg, Sweden
- Department of Drug Treatment, Region Västra Götaland, Sahlgrenska University Hospital, SE-41345 Gothenburg, Sweden
| |
Collapse
|
17
|
Kieler M, Prammer LS, Heller G, Hofmann M, Sperger S, Hanetseder D, Niederreiter B, Komljenovic A, Klavins K, Köcher T, Brunner JS, Stanic I, Oberbichler L, Korosec A, Vogel A, Kerndl M, Hromadová D, Musiejovsky L, Hajto A, Dobrijevic A, Piwonka T, Haschemi A, Miller A, Georgel P, Marolt Presen D, Grillari J, Hayer S, Auger JP, Krönke G, Sharif O, Aletaha D, Schabbauer G, Blüml S. Itaconate is a metabolic regulator of bone formation in homeostasis and arthritis. Ann Rheum Dis 2024; 83:1465-1479. [PMID: 38986577 PMCID: PMC11503170 DOI: 10.1136/ard-2023-224898] [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: 08/22/2023] [Accepted: 06/19/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVES Bone remodelling is a highly dynamic process dependent on the precise coordination of osteoblasts and haematopoietic-cell derived osteoclasts. Changes in core metabolic pathways during osteoclastogenesis, however, are largely unexplored and it is unknown whether and how these processes are involved in bone homeostasis. METHODS We metabolically and transcriptionally profiled cells during osteoclast and osteoblast generation. Individual gene expression was characterised by quantitative PCR and western blot. Osteoblast function was assessed by Alizarin red staining. immunoresponsive gene 1 (Irg1)-deficient mice were used in various inflammatory or non-inflammatory models of bone loss. Tissue gene expression was analysed by RNA in situ hybridisation. RESULTS We show that during differentiation preosteoclasts rearrange their tricarboxylic acid cycle, a process crucially depending on both glucose and glutamine. This rearrangement is characterised by the induction of Irg1 and production of itaconate, which accumulates intracellularly and extracellularly. While the IRG1-itaconate axis is dispensable for osteoclast generation in vitro and in vivo, we demonstrate that itaconate stimulates osteoblasts by accelerating osteogenic differentiation in both human and murine cells. This enhanced osteogenic differentiation is accompanied by reduced proliferation and altered metabolism. Additionally, supplementation of itaconate increases bone formation by boosting osteoblast activity in mice. Conversely, Irg1-deficient mice exhibit decreased bone mass and have reduced osteoproliferative lesions in experimental arthritis. CONCLUSION In summary, we identify itaconate, generated as a result of the metabolic rewiring during osteoclast differentiation, as a previously unrecognised regulator of osteoblasts.
Collapse
Affiliation(s)
- Markus Kieler
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Leona Sophia Prammer
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Gerwin Heller
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Melanie Hofmann
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Simon Sperger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Dominik Hanetseder
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Birgit Niederreiter
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Andrea Komljenovic
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Christian Doppler Laboratory for Immunometabolism and Systems Biology of Obesity-Related Diseases (InSpiReD), Vienna, Austria
| | - Kristaps Klavins
- Institute of General Chemical Engineering, Riga Technical University, Riga, Latvia
| | - Thomas Köcher
- Vienna BioCenter Core Facilities, Campus-Vienna-BioCenter 1, Vienna, Austria
| | - Julia Stefanie Brunner
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Irena Stanic
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
| | - Laura Oberbichler
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ana Korosec
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Christian Doppler Laboratory for Immunometabolism and Systems Biology of Obesity-Related Diseases (InSpiReD), Vienna, Austria
| | - Andrea Vogel
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
| | - Martina Kerndl
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
| | - Dominika Hromadová
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
| | - Laszlo Musiejovsky
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
| | - Alexander Hajto
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
| | - Anja Dobrijevic
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Christian Doppler Laboratory for Immunometabolism and Systems Biology of Obesity-Related Diseases (InSpiReD), Vienna, Austria
| | - Tina Piwonka
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Wien, Austria
| | - Anne Miller
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Philippe Georgel
- INSERM UMR_S 1109, Fédération de Médecine Translationnelle (FMTS), Université de Strasbourg, Centre de Recherche en Immunologie et Hématologie, 1 Place de l’Hôpital, Strasbourg Cedex, France
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Wien, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Silvia Hayer
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Jean-Philippe Auger
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Omar Sharif
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Christian Doppler Laboratory for Immunometabolism and Systems Biology of Obesity-Related Diseases (InSpiReD), Vienna, Austria
| | - Daniel Aletaha
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Gernot Schabbauer
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University of Vienna, Wien, Vienna, Austria
- Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Stephan Blüml
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| |
Collapse
|
18
|
Li Q, Wang J, Zhao C. From Genomics to Metabolomics: Molecular Insights into Osteoporosis for Enhanced Diagnostic and Therapeutic Approaches. Biomedicines 2024; 12:2389. [PMID: 39457701 PMCID: PMC11505085 DOI: 10.3390/biomedicines12102389] [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: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Osteoporosis (OP) is a prevalent skeletal disorder characterized by decreased bone mineral density (BMD) and increased fracture risk. The advancements in omics technologies-genomics, transcriptomics, proteomics, and metabolomics-have provided significant insights into the molecular mechanisms driving OP. These technologies offer critical perspectives on genetic predispositions, gene expression regulation, protein signatures, and metabolic alterations, enabling the identification of novel biomarkers for diagnosis and therapeutic targets. This review underscores the potential of these multi-omics approaches to bridge the gap between basic research and clinical applications, paving the way for precision medicine in OP management. By integrating these technologies, researchers can contribute to improved diagnostics, preventative strategies, and treatments for patients suffering from OP and related conditions.
Collapse
Affiliation(s)
- Qingmei Li
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Jihan Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
| | - Congzhe Zhao
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| |
Collapse
|
19
|
Deng X, Wu X, Sun Z, Liu Q, Yuan G. Associations between new obesity indices and abnormal bone density in type 2 diabetes mellitus patients. Osteoporos Int 2024; 35:1807-1815. [PMID: 38965122 DOI: 10.1007/s00198-024-07163-9] [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] [Received: 01/15/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024]
Abstract
The clinical data analysis found that, compared with the traditional obesity index, the waist-weight ratio (WWR) has more advantages in predicting abnormal bone mineral density in subjects with type 2 diabetes. WWR may serve as a new predictive indicator for osteoporosis in T2DM patients. PURPOSE This study was designed to explore the correlation between obesity-related indices and bone mineral density (BMD) and its influencing factors in type 2 diabetes mellitus (T2DM) patients. METHODS A total of 528 patients with type 2 diabetes were recruited. Glucose tolerance, insulin stimulation, and blood biochemical tests were conducted on all participants. All subjects underwent dual-energy X-ray bone density testing and were grouped based on the bone density results. RESULTS Compared with those in the normal BMD group, the waist-to-body weight ratio (WWR) and weight-adjusted-waist index (WWI) in the osteopenia and osteoporosis groups were significantly greater, while body mass index (BMI) was significantly lower (P < 0.05). The logistic regression results showed that the WWR, WWI, and BMI were independently correlated with abnormal BMD in T2DM patients (P < 0.05). WWR and the WWI were negatively correlated with the T-value of bone density in various parts of the body, while BMI was positively correlated with the T-value of bone density (P < 0.05). The area under the working characteristic curve (AUC) for T2DM patients with abnormal bone mass predicted by the WWR [0.806, 95% CI = (0.770-0.843), P < 0.001] was greater than that for patients with other obesity indicators, such as the WWI and BMI. CONCLUSION We found a positive correlation between the WWR and bone density in T2DM patients. Compared with other obesity indicators, such as BMI and WWI, the WWR has a stronger discriminative ability for T2DM patients with abnormal bone density. Therefore, more attention should be given to the WWR in T2DM patients.
Collapse
Affiliation(s)
- Xia Deng
- Department of Endocrinology and Metabolism, Institute of Endocrine and Metabolic Diseases, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xunan Wu
- Department of Endocrinology and Metabolism, Institute of Endocrine and Metabolic Diseases, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ziyan Sun
- Department of Endocrinology and Metabolism, Institute of Endocrine and Metabolic Diseases, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiaoyan Liu
- Department of Endocrinology and Metabolism, Institute of Endocrine and Metabolic Diseases, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolism, Institute of Endocrine and Metabolic Diseases, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China.
- Endocrine Research Institute, The affiliated hospital of jiangsu university, Zhenjiang, China.
| |
Collapse
|
20
|
Zhang B, Shang P. Protocol for evaluating the effects of large gradient high magnetic fields on osteocyte function. STAR Protoc 2024; 5:103186. [PMID: 39003746 PMCID: PMC11284420 DOI: 10.1016/j.xpro.2024.103186] [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: 02/07/2024] [Revised: 05/04/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Osteocytes are the main mechanosensory cells and the primary regulators of bone metabolic homeostasis. Here, we present a protocol for evaluating the effects of the large gradient high magnetic field (LG-HMF) on osteocyte function. We describe steps for establishing a corresponding cell culture system in the LG-HMF generated by a superconducting magnet. We then detail procedures for using this cell culture system to study the effects of magnetic forces on the structure and function of murine long bone osteocyte Y4 cells. For complete details on the use and execution of this protocol, please refer to Zhang et al.1.
Collapse
Affiliation(s)
- Bin Zhang
- School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
21
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
22
|
Suh J, Lee YS. The multifaceted roles of mitochondria in osteoblasts: from energy production to mitochondrial-derived vesicle secretion. J Bone Miner Res 2024; 39:1205-1214. [PMID: 38907370 PMCID: PMC11371665 DOI: 10.1093/jbmr/zjae088] [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: 01/12/2024] [Revised: 05/03/2024] [Indexed: 06/24/2024]
Abstract
Mitochondria in osteoblasts have been demonstrated to play multiple crucial functions in bone formation from intracellular adenosine triphosphate production to extracellular secretion of mitochondrial components. The present review explores the current knowledge about mitochondrial biology in osteoblasts, including mitochondrial biogenesis, bioenergetics, oxidative stress generation, and dynamic changes in morphology. Special attention is given to recent findings, including mitochondrial donut formation in osteoblasts, which actively generates mitochondrial-derived vesicles (MDVs), followed by extracellular secretion of small mitochondria and MDVs. We also discuss the therapeutic effects of targeting osteoblast mitochondria, highlighting their potential applications in improving bone health.
Collapse
Affiliation(s)
- Joonho Suh
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
23
|
Kolipaka R, Magesh I, Bharathy MA, Karthik S, Saranya I, Selvamurugan N. A potential function for MicroRNA-124 in normal and pathological bone conditions. Noncoding RNA Res 2024; 9:687-694. [PMID: 38577015 PMCID: PMC10990750 DOI: 10.1016/j.ncrna.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 04/06/2024] Open
Abstract
Cells produce short single-stranded non-coding RNAs (ncRNAs) called microRNAs (miRNAs), which actively regulate gene expression at the posttranscriptional level. Several miRNAs have been observed to exert significant impacts on bone health and bone-related disorders. One of these, miR-124, is observed in bone microenvironments and is conserved across species. It affects bone cell growth and differentiation by activating different transcription factors and signaling pathways. In-depth functional analyses of miR-124 have revealed several physiological and pathological roles exerted through interactions with other ncRNAs. Deciphering these RNA-mediated signaling networks and pathways is essential for understanding the potential impacts of dysregulated miRNA functions on bone biology. In this review, we aim to provide a comprehensive analysis of miR-124's involvement in bone physiology and pathology. We highlight the importance of miR-124 in controlling transcription factors and signaling pathways that promote bone growth. This review reveals therapeutic implications for the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Rushil Kolipaka
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Induja Magesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - M.R. Ashok Bharathy
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - S. Karthik
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - I. Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - N. Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
24
|
Dzubanova M, Benova A, Ferencakova M, Coupeau R, Tencerova M. Nutrition and Bone Marrow Adiposity in Relation to Bone Health. Physiol Res 2024; 73:S107-S138. [PMID: 38752771 PMCID: PMC11412336 DOI: 10.33549/physiolres.935293] [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: 12/02/2023] [Accepted: 04/29/2024] [Indexed: 09/04/2024] Open
Abstract
Bone remodeling is energetically demanding process. Energy coming from nutrients present in the diet contributes to function of different cell type including osteoblasts, osteocytes and osteoclasts in bone marrow participating in bone homeostasis. With aging, obesity and osteoporosis the function of key building blocks, bone marrow stromal cells (BMSCs), changes towards higher accumulation of bone marrow adipose tissue (BMAT) and decreased bone mass, which is affected by diet and sex dimorphism. Men and women have unique nutritional needs based on physiological and hormonal changes across the life span. However, the exact molecular mechanisms behind these pathophysiological conditions in bone are not well-known. In this review, we focus on bone and BMAT physiology in men and women and how this approach has been taken by animal studies. Furthermore, we discuss the different diet interventions and impact on bone and BMAT in respect to sex differences. We also discuss the future perspective on precision nutrition with a consideration of sex-based differences which could bring better understanding of the diet intervention in bone health and weight management.
Collapse
Affiliation(s)
- M Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague 4, Czech Republic.
| | | | | | | | | |
Collapse
|
25
|
Sayeed UB, Akhtar E, Roy AK, Akter S, von Ehrenstein OS, Raqib R, Wagatsuma Y. Fetal femur length and risk of diabetes in adolescence: a prospective cohort study. Trop Med Health 2024; 52:44. [PMID: 38951934 PMCID: PMC11218328 DOI: 10.1186/s41182-024-00611-6] [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: 04/10/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Diabetes is more apparent in adulthood but may be dormant in childhood and originates during early fetal development. In fetal biometry, femur length (FL) is crucial for assessing fetal growth and development. This study aimed to assess potential associations between fetal femur growth and prediabetic biomarkers in Bangladeshi children. METHODS A cohort study embedded in a population-based maternal food and micronutrient supplementation (MINIMat) trial was conducted in Matlab, Bangladesh. The children in the cohort were followed up until 15 years of age. In the original trial, pregnancy was confirmed by ultrasound before 13 gestational weeks (GWs). Afterward, ultrasound assessments were performed at 14, 19, and 30 GWs. FL was measured from one end to the other, capturing a complete femoral image. The FL was standardized by GW, and a z-score was calculated. FBG and HbA1c levels were determined in plasma and whole blood, and the triglyceride-glucose index, a biomarker of insulin resistance, was calculated as Ln [fasting triglycerides (mg/dl) × fasting glucose (mg/dl)/2]. Multivariable linear regression analysis using a generalized linear model was performed to estimate the effects of FL at 14, 19 and 30 GWs on prediabetic biomarkers at 9 and 15 years of age. Maternal micronutrient and food supplementation group, parity, child sex, and BMI at 9 years or 15 years were included as covariates. RESULTS A total of 1.2% (6/515) of the participants had impaired fasting glucose during preadolescence, which increased to 3.5% (15/433) during adolescence. At 9 years, 6.3% (32/508) of the participants had elevated HbA1c%, which increased to 28% (120/431) at 15 years. Additionally, the TyG index increased from 9.5% (49/515) (during preadolescence) to 13% (56/433) (during adolescence). A one standard deviation decrease in FL at 14 and 19 GWs was associated with increased FBG (β = - 0.44 [- 0.88, - 0.004], P = 0.048; β = - 0.59 [- 1.12, - 0.05], P = 0.031) and HbA1c (β = - 0.01; [- 0.03, -0.005], P = 0.007; β = - 0.01 [- 0.03, - 0.003], P = 0.018) levels at 15 years. FL was not associated with diabetic biomarkers at 9 years. CONCLUSION Mid-trimester impaired femur growth may be associated with elevated prediabetic biomarkers in Bangladeshi adolescents.
Collapse
Affiliation(s)
- Urme Binte Sayeed
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Evana Akhtar
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Anjan Kumar Roy
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Sharmin Akter
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Ondine S von Ehrenstein
- Departments of Community Health Sciences and Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Rubhana Raqib
- Nutrition Research Division, icddr, b, Dhaka, 1212, Bangladesh
| | - Yukiko Wagatsuma
- Faculty of Medicine, Department of Clinical Trials and Clinical Epidemiology, University of Tsukuba, Tsukuba, Ibaraki, 805-3575, Japan
| |
Collapse
|
26
|
van Brakel F, Zhao Y, van der Eerden BC. Fueling recovery: The importance of energy coupling between angiogenesis and osteogenesis during fracture healing. Bone Rep 2024; 21:101757. [PMID: 38577251 PMCID: PMC10990718 DOI: 10.1016/j.bonr.2024.101757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 04/06/2024] Open
Abstract
Approximately half of bone fractures that do not heal properly (non-union) can be accounted to insufficient angiogenesis. The processes of angiogenesis and osteogenesis are spatiotemporally regulated in the complex process of fracture healing that requires a substantial amount of energy. It is thought that a metabolic coupling between angiogenesis and osteogenesis is essential for successful healing. However, how this coupling is achieved remains to be largely elucidated. Here, we will discuss the most recent evidence from literature pointing towards a metabolic coupling between angiogenesis and osteogenesis. We will describe the metabolic profiles of the cell types involved during fracture healing as well as secreted products in the bone microenvironment (such as lactate and nitric oxide) as possible key players in this metabolic crosstalk.
Collapse
Affiliation(s)
- Fleur van Brakel
- Calcium and Bone Metabolism Laboratory, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yudong Zhao
- Calcium and Bone Metabolism Laboratory, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bram C.J. van der Eerden
- Calcium and Bone Metabolism Laboratory, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
27
|
Zhou F, Wang Z, Zhang G, Wu Y, Xiong Y. Immunosenescence and inflammaging: Conspiracies against alveolar bone turnover. Oral Dis 2024; 30:1806-1817. [PMID: 37288702 DOI: 10.1111/odi.14642] [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: 01/25/2023] [Revised: 05/11/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE Inflammaging and immunosenescence are characteristics of senescent immune system alterations. This review provides insights into inflammaging and immunosenescence in periodontitis and focuses on the innerlink of inflammaging and immunosenescence in alveolar bone turnover from a perspective of cell-cell interaction. METHODS This review is conducted by a narrative approach to discuss the effect of inflammaging and immunosenescence in aging-related alveolar bone loss. A comprehensive literature research in PubMed and Google was applied to identify reports in English. RESULTS Inflammaging is concerned with abnormal M1 polarization and increasing circulating inflammatory cytokines, while immunosenescence involves reduced infection and vaccine responses, depressed antimicrobial function, and infiltration of aged B cells and memory T cells. TLR-mediated inflammaging and altered adaptive immunity significantly affect alveolar bone turnover and aggravate aging-related alveolar bone loss. Besides, energy consumption also plays a vital role in aged immune and skeletal system of periodontitis. CONCLUSIONS Senescent immune system exerts a significant function in aging-related alveolar bone loss. Inflammaging and immunosenescence interact functionally and mechanistically, which affects alveolar bone turnover. Therefore, further clinical treatment strategies targeting alveolar bone loss could be based on the specific molecular mechanism connecting inflammaging, immunosenescence, and alveolar bone turnover.
Collapse
Affiliation(s)
- Feng Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhanqi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guorui Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yingying Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Gao F, Hu Q, Chen W, Li J, Qi C, Yan Y, Qian C, Wan M, Ficke J, Zheng J, Cao X. Brain regulates weight bearing bone through PGE2 skeletal interoception: implication of ankle osteoarthritis and pain. Bone Res 2024; 12:16. [PMID: 38443372 PMCID: PMC10914853 DOI: 10.1038/s41413-024-00316-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 03/07/2024] Open
Abstract
Bone is a mechanosensitive tissue and undergoes constant remodeling to adapt to the mechanical loading environment. However, it is unclear whether the signals of bone cells in response to mechanical stress are processed and interpreted in the brain. In this study, we found that the hypothalamus of the brain regulates bone remodeling and structure by perceiving bone prostaglandin E2 (PGE2) concentration in response to mechanical loading. Bone PGE2 levels are in proportion to their weight bearing. When weight bearing changes in the tail-suspension mice, the PGE2 concentrations in bones change in line with their weight bearing changes. Deletion of cyclooxygenase-2 (COX2) in the osteoblast lineage cells or knockout of receptor 4 (EP4) in sensory nerve blunts bone formation in response to mechanical loading. Moreover, knockout of TrkA in sensory nerve also significantly reduces mechanical load-induced bone formation. Moreover, mechanical loading induces cAMP-response element binding protein (CREB) phosphorylation in the hypothalamic arcuate nucleus (ARC) to inhibit sympathetic tyrosine hydroxylase (TH) expression in the paraventricular nucleus (PVN) for osteogenesis. Finally, we show that elevated PGE2 is associated with ankle osteoarthritis (AOA) and pain. Together, our data demonstrate that in response to mechanical loading, skeletal interoception occurs in the form of hypothalamic processing of PGE2-driven peripheral signaling to maintain physiologic bone homeostasis, while chronically elevated PGE2 can be sensed as pain during AOA and implication of potential treatment.
Collapse
Affiliation(s)
- Feng Gao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Qimiao Hu
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Wenwei Chen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Jilong Li
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Cheng Qi
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Yiwen Yan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Cheng Qian
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Mei Wan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - James Ficke
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Junying Zheng
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.
| |
Collapse
|
29
|
Liu J, Bao X, Huang J, Chen R, Tan Y, Zhang Z, Xiao B, Kong F, Gu C, Du J, Wang H, Qi J, Tan J, Ma D, Shi C, Xu G. TMEM135 maintains the equilibrium of osteogenesis and adipogenesis by regulating mitochondrial dynamics. Metabolism 2024; 152:155767. [PMID: 38154611 DOI: 10.1016/j.metabol.2023.155767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Disturbance in the differentiation process of bone marrow mesenchymal stem cells (BMSCs) leads to osteoporosis. Mitochondrial dynamics plays a pivotal role in the metabolism and differentiation of BMSCs. However, the mechanisms underlying mitochondrial dynamics and their impact on the differentiation equilibrium of BMSCs remain unclear. METHODS We investigated the mitochondrial morphology and markers related to mitochondrial dynamics during BMSCs osteogenic and adipogenic differentiation. Bioinformatics was used to screen potential genes regulating BMSCs differentiation through mitochondrial dynamics. Subsequently, we evaluated the impact of Transmembrane protein 135 (TMEM135) deficiency on bone homeostasis by comparing Tmem135 knockout mice with their littermates. The mechanism of TMEM135 in mitochondrial dynamics and BMSCs differentiation was also investigated in vivo and in vitro. RESULTS Distinct changes in mitochondrial morphology were observed between osteogenic and adipogenic differentiation of BMSCs, manifesting as fission in the late stage of osteogenesis and fusion in adipogenesis. Additionally, we revealed that TMEM135, a modulator of mitochondrial dynamics, played a functional role in regulating the equilibrium between adipogenesis and osteogenesis. The TMEM135 deficiency impaired mitochondrial fission and disrupted crucial mitochondrial energy metabolism during osteogenesis. Tmem135 knockout mice showed osteoporotic phenotype, characterized by reduced osteogenesis and increased adipogenesis. Mechanistically, TMEM135 maintained intracellular calcium ion homeostasis and facilitated the dephosphorylation of dynamic-related protein 1 at Serine 637 in BMSCs. CONCLUSIONS Our findings underscore the significant role of TMEM135 as a modulator in orchestrating the differentiation trajectory of BMSCs and promoting a shift in mitochondrial dynamics toward fission. This ultimately contributes to the osteogenesis process. This work has provided promising biological targets for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Jia Liu
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Xiaogang Bao
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Jian Huang
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Rukun Chen
- Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Yixuan Tan
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Zheng Zhang
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Bing Xiao
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Fanqi Kong
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Changjiang Gu
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Jianhang Du
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Haotian Wang
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Junqiang Qi
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China
| | - Junming Tan
- Department of Orthopedics, The 72nd Army Hospital of the People's Liberation Army, Huzhou 313099, PR China
| | - Duan Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China.
| | - Changgui Shi
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China.
| | - Guohua Xu
- Department of Orthopedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, PR China.
| |
Collapse
|
30
|
Zhang Y, Kong Y, Zhang W, He J, Zhang Z, Cai Y, Zhao Y, Xu Q. METTL3 promotes osteoblast ribosome biogenesis and alleviates periodontitis. Clin Epigenetics 2024; 16:18. [PMID: 38267969 PMCID: PMC10809637 DOI: 10.1186/s13148-024-01628-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Periodontitis is a highly prevalent oral disease characterized by bacterium-induced periodontal inflammation and alveolar bone destruction. Osteoblast function is impaired in periodontitis with a global proteome change. METTL3 is the pivotal methyltransferase of N6-methyladenosine (m6A) that is recently proved to exert a crucial role in osteoblast differentiation. This study aims to investigate the role of METTL3 in osteoblast ribosome biogenesis in periodontitis progression. RESULTS METTL3 was knocked down in osteoblasts, and the downregulated genes were enriched in ribosome and translation. METTL3 knockdown inhibited ribosome biogenesis and oxidative phosphorylation in LPS-stimulated osteoblasts, whereas METTL3 overexpression facilitated ribosomal and mitochondrial function. Mechanistically, METTL3 mediated osteoblast biological behaviors by activating Wnt/β-catenin/c-Myc signaling. METTL3 depletion enhanced the mRNA expression and stability of Dkk3 and Sostdc1 via YTHDF2. In periodontitis mice, METTL3 inhibitor SAH promoted alveolar bone loss and local inflammatory status, which were partially rescued by Wnt/β-catenin pathway activator CHIR-99021 HCl. CONCLUSIONS METTL3 promoted ribosome biogenesis and oxidative phosphorylation by activating Wnt/β-catenin/c-Myc signaling in LPS-treated osteoblasts and alleviated the inflammatory alveolar bone destruction in periodontitis mice.
Collapse
Affiliation(s)
- Yiwen Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Ling Yuan Xi Road, Guangzhou, 510055, China
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yiping Kong
- Changsha Stomatological Hospital, Hunan University of Chinese Medicine, Changsha, 410004, China
| | - Wenjie Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Ling Yuan Xi Road, Guangzhou, 510055, China
| | - Jinlin He
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Ling Yuan Xi Road, Guangzhou, 510055, China
| | - Zhanqi Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Ling Yuan Xi Road, Guangzhou, 510055, China
| | - Yongjie Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Ling Yuan Xi Road, Guangzhou, 510055, China
| | - Yiqing Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Ling Yuan Xi Road, Guangzhou, 510055, China
| | - Qiong Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, 56 Ling Yuan Xi Road, Guangzhou, 510055, China.
| |
Collapse
|
31
|
Ledesma-Colunga MG, Passin V, Lademann F, Hofbauer LC, Rauner M. Novel Insights into Osteoclast Energy Metabolism. Curr Osteoporos Rep 2023; 21:660-669. [PMID: 37816910 PMCID: PMC10724336 DOI: 10.1007/s11914-023-00825-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2023] [Indexed: 10/12/2023]
Abstract
PURPOSE OF REVIEW Osteoclasts are crucial for the dynamic remodeling of bone as they resorb old and damaged bone, making space for new bone. Metabolic reprogramming in these cells not only supports phenotypic changes, but also provides the necessary energy for their highly energy-consuming activity, bone resorption. In this review, we highlight recent developments in our understanding of the metabolic adaptations that influence osteoclast behavior and the overall remodeling of bone tissue. RECENT FINDINGS Osteoclasts undergo metabolic reprogramming to meet the energy demands during their transition from precursor cells to fully mature bone-resorbing osteoclasts. Recent research has made considerable progress in pinpointing crucial metabolic adaptations and checkpoint proteins in this process. Notably, glucose metabolism, mitochondrial biogenesis, and oxidative respiration were identified as essential pathways involved in osteoclast differentiation, cytoskeletal organization, and resorptive activity. Furthermore, the interaction between these pathways and amino acid and lipid metabolism adds to the complexity of the process. These interconnected processes can function as diverse fuel sources or have independent regulatory effects, significantly influencing osteoclast function. Energy metabolism in osteoclasts involves various substrates and pathways to meet the energetic requirements of osteoclasts throughout their maturation stages. This understanding of osteoclast biology may provide valuable insights for modulating osteoclast activity during the pathogenesis of bone-related disorders and may pave the way for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Maria G Ledesma-Colunga
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Vanessa Passin
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Franziska Lademann
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, 01307, Dresden, Germany.
| |
Collapse
|
32
|
Liu Z, Wang R, Liu W, Liu Y, Feng X, Zhao F, Chen P, Shao L, Rong M. Recent advances in the application and biological mechanism of silicon nitride osteogenic properties: a review. Biomater Sci 2023; 11:7003-7017. [PMID: 37718623 DOI: 10.1039/d3bm00877k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Silicon nitride, an emerging bioceramic material, is highly sought after in the biomedical industry due to its osteogenesis-promoting properties, which are a result of its unique surface chemistry and excellent mechanical properties. Currently, it is used in clinics as an orthopedic implant material. The osteogenesis-promoting properties of silicon nitride are manifested in its contribution to the formation of a local osteogenic microenvironment, wherein silicon nitride and its hydrolysis products influence osteogenesis by modulating the biological behaviors of the constituents of the osteogenic microenvironment. In particular, silicon nitride regulates redox signaling, cellular autophagy, glycolysis, and bone mineralization in cells involved in bone formation via several mechanisms. Moreover, it may also promote osteogenesis by influencing immune regulation and angiogenesis. In addition, the wettability, surface morphology, and charge of silicon nitride play crucial roles in regulating its osteogenesis-promoting properties. However, as a bioceramic material, the molding process of silicon nitride needs to be optimized, and its osteogenic mechanism must be further investigated. Herein, we summarize the impact of the molding process of silicon nitride on its osteogenic properties and clinical applications. In addition, the mechanisms of silicon nitride in promoting osteogenesis are discussed, followed by a summary of the current gaps in silicon nitride mechanism research. This review, therefore, aims to provide novel ideas for the future development and applications of silicon nitride.
Collapse
Affiliation(s)
- Ziyi Liu
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Ruijie Wang
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Wenjing Liu
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Yushan Liu
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Xiaoli Feng
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Fujian Zhao
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Pei Chen
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| | - Mingdeng Rong
- Stomatological Hospital, Southern Medical University, Jiangnan Avenue 366, Guangzhou 510280, China.
| |
Collapse
|
33
|
Chen S, Wang S, Ding S, Zhang C. Evaluation of Tibial Hemodynamic Response to Glucose Tolerance Test in Young Healthy Males and Females. Nutrients 2023; 15:4062. [PMID: 37764845 PMCID: PMC10535503 DOI: 10.3390/nu15184062] [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: 08/09/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The relationship between glucose metabolism and bone health remains underexplored despite its clinical relevance. This study utilized the oral glucose tolerance test (OGTT) and near-infrared spectroscopy (NIRS) to probe gender-specific disparities in tibial hemodynamic responses among young healthy adults. Twenty-eight healthy participants (14 males) aged 18-28 years old were recruited for this study. After ingesting a 75 g glucose solution, tibial hemodynamic responses were captured using NIRS in combination with a 5 min ischemic reperfusion technique, both before and at 30 min intervals for two hours post-glucose ingestion. Parameters measured included oxidative metabolic rate (via tissue saturation index [TSI]), immediate recovery slope after occlusion release (TSI10), and total recovery magnitude (ΔTSI). Post-glucose ingestion, both genders demonstrated a surge in blood glucose concentrations at every time point compared to baseline (p < 0.001, 0.002, 0.009, and 0.039 for males; p < 0.001, < 0.001, = 0.002, and 0.017 for females). Baseline tibial metabolic rate, TSI10, and ΔTSI did not significantly differ between males and females (p = 0.734, 0.839, and 0.164, respectively), with no discernible temporal effects in any hemodynamic parameters within each gender (p = 0.864, 0.308, and 0.399, respectively, for males; p = 0.973, 0.453, and 0.137, respectively, for females). We found comparable tibial hemodynamic responses to OGTT between genders. This study demonstrated the utility of NIRS in evaluating tibial hemodynamic responses to glucose ingestion through OGTT, enriching our understanding of the body's metabolic responses to glucose intake.
Collapse
Affiliation(s)
- Si Chen
- School of Physical Education and Sport, Central China Normal University, Wuhan 430079, China; (S.C.); (S.D.)
| | - Shubo Wang
- Globus Medical Inc., Audubon, PA 19403, USA;
| | - Shuqiao Ding
- School of Physical Education and Sport, Central China Normal University, Wuhan 430079, China; (S.C.); (S.D.)
| | - Chuan Zhang
- School of Physical Education and Sport, Central China Normal University, Wuhan 430079, China; (S.C.); (S.D.)
| |
Collapse
|
34
|
Gao F, Hu Q, Qi C, Wan M, Ficke J, Zheng J, Cao X. Mechanical loading-induced change of bone homeostasis is mediated by PGE2-driven hypothalamic interoception. RESEARCH SQUARE 2023:rs.3.rs-3325498. [PMID: 37790467 PMCID: PMC10543368 DOI: 10.21203/rs.3.rs-3325498/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Bone is a mechanosensitive tissue and undergoes constant remodeling to adapt to the mechanical loading environment. However, it is unclear whether the signals of bone cells in response to mechanical stress are processed and interpreted in the brain. In this study, we found that the hypothalamus of the brain regulates bone remodeling and structure by perceiving bone PGE2 concentration in response to mechanical loading. Bone PGE2 levels are in proportion to their weight bearing. When weight bearing changes in the tail-suspension mice, the PGE2 concentrations in bones change in line with their weight bearing changes. Deletion of Cox2 or Pge2 in the osteoblast lineage cells or knockout Ep4 in sensory nerve blunts bone formation in response to mechanical loading. And sensory denervation also significantly reduces mechanical load-induced bone formation. Moreover, mechanical loading induces CREB phosphorylation in the hypothalamic ARC region to inhibit sympathetic TH expression in the PVN for osteogenesis. Finally, we show that elevated PGE2 is associated with ankle osteoarthritis (AOA) and pain. Together, our data demonstrate that in response to mechanical loading, skeletal interoception occurs in the form of hypothalamic processing of PGE2-driven peripheral signaling to maintain physiologic bone homeostasis, while chronically elevated PGE2 can be sensed as pain during AOA and implication of potential treatment.
Collapse
Affiliation(s)
- Feng Gao
- Department of Orthopedic Surgery and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Qimiao Hu
- Department of Orthopedic Surgery and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Cheng Qi
- Department of Orthopedic Surgery and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Mei Wan
- Department of Orthopedic Surgery and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - James Ficke
- Department of Orthopedic Surgery and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Junying Zheng
- Department of Orthopedic Surgery and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Xu Cao
- Department of Orthopedic Surgery and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
35
|
Lu W, Duan Y, Li K, Qiu J, Cheng Z. Glucose uptake and distribution across the human skeleton using state-of-the-art total-body PET/CT. Bone Res 2023; 11:36. [PMID: 37407553 DOI: 10.1038/s41413-023-00268-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/25/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
A growing number of studies have demonstrated that the skeleton is an endocrine organ that is involved in glucose metabolism and plays a significant role in human glucose homeostasis. However, there is still a limited understanding of the in vivo glucose uptake and distribution across the human skeleton. To address this issue, we aimed to elucidate the detailed profile of glucose uptake across the skeleton using a total-body positron emission tomography (PET) scanner. A total of 41 healthy participants were recruited. Two of them received a 1-hour dynamic total-body 18F-fluorodeoxyglucose (18F-FDG) PET scan, and all of them received a 10-minute static total-body 18F-FDG PET scan. The net influx rate (Ki) and standardized uptake value normalized by lean body mass (SUL) were calculated as indicators of glucose uptake from the dynamic and static PET data, respectively. The results showed that the vertebrae, hip bone and skull had relatively high Ki and SUL values compared with metabolic organs such as the liver. Both the Ki and SUL were higher in the epiphyseal, metaphyseal and cortical regions of long bones. Moreover, trends associated with age and overweight with glucose uptake (SULmax and SULmean) in bones were uncovered. Overall, these results indicate that the skeleton is a site with significant glucose uptake, and skeletal glucose uptake can be affected by age and dysregulated metabolism.
Collapse
Affiliation(s)
- Weizhao Lu
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, China
| | - Yanhua Duan
- Department of PET-CT, the First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital Affiliated with Shandong University, Jinan, 250014, China
| | - Kun Li
- Department of PET-CT, the First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital Affiliated with Shandong University, Jinan, 250014, China
| | - Jianfeng Qiu
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, China.
| | - Zhaoping Cheng
- Department of PET-CT, the First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital Affiliated with Shandong University, Jinan, 250014, China.
| |
Collapse
|
36
|
Zhang Q, Liu J, Lin H, Lin B, Zhu M, Li M. Glucose metabolism reprogramming promotes immune escape of hepatocellular carcinoma cells. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:519-536. [PMID: 37455832 PMCID: PMC10344893 DOI: 10.37349/etat.2023.00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 07/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a complex process that plays an important role in its progression. Abnormal glucose metabolism in HCC cells can meet the nutrients required for the occurrence and development of liver cancer, better adapt to changes in the surrounding microenvironment, and escape the attack of the immune system on the tumor. There is a close relationship between reprogramming of glucose metabolism and immune escape. This article reviews the current status and progress of glucose metabolism reprogramming in promoting immune escape in liver cancer, aiming to provide new strategies for clinical immunotherapy of liver cancer.
Collapse
Affiliation(s)
- Qiuyue Zhang
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Jinchen Liu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Haifeng Lin
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical College, Haikou 570216, Hainan Province, China
| | - Bo Lin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, Hainan Province, China
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical College, Haikou 570216, Hainan Province, China
- Institution of Tumor, Hainan Medical College, Haikou 570102, Hainan Province, China
| |
Collapse
|
37
|
Wang W, Chen ZY, Lv FY, Tu M, Guo XL. Apolipoprotein A1 is associated with osteocalcin and bone mineral density rather than high-density lipoprotein cholesterol in Chinese postmenopausal women with type 2 diabetes mellitus. Front Med (Lausanne) 2023; 10:1182866. [PMID: 37396919 PMCID: PMC10308019 DOI: 10.3389/fmed.2023.1182866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Objective Disturbances in high-density lipoprotein cholesterol (HDL-c) metabolic pathways can affect bone metabolism, which may rely on the particle function of apolipoprotein rather than HDL-c levels. This study aimed to evaluate the correlation of serum HDL-c and apolipoprotein A1 (APOA1) with bone metabolism in Chinese postmenopausal women with type 2 diabetes mellitus (T2DM). Method A total of 1,053 participants with complete data were enrolled and separated into three groups based on the HDL-c and APOA1 tertiles. The trained reviewer collected demographic and anthropometric information. Bone turnover markers (BTMs) were determined by standard methods. Bone mineral density (BMD) was measured by dual-energy x-ray absorptiometry. Results Overall, the prevalence of osteoporosis was 29.7%. Groups with higher APOA1 have a remarkably more elevated level of osteocalcin (OC), L1-L4 BMD, and T-score across the APOA1 tertiles. APOA1 presented a positive correlation with OC (r = 0.194, p < 0.001), L1-L4 BMD (r = 0.165, p < 0.001), and T-score (r = 0.153, p < 0.001) rather than HDL-c. Meanwhile, APOA1 remained independently associated with OC (β = 0.126, p < 0.001), L1-L4 BMD (β = 0.181, p < 0.001), and T-score (β = 0.180, p < 0.001) after adjustment for confounding factors. APOA1 is also shown to be independently correlated with osteoporosis after adjustment for confounding factors, and the OR (95%CI) was 0.851 (0.784-0.924). In contrast, there was no significant association between HDL-c and osteoporosis. Furthermore, APOA1 seemed to have the largest areas under the curve (AUC) for osteoporosis. The AUC (95% CI) of APOA1 identifying osteoporosis was 0.615 (0.577-0.652). The optimal cut-off value of APOA1 was 0.89 g/L (sensitivity: 56.5%, specificity: 67.9%). Conclusion APOA1 is independently associated with OC, L1-L4 BMD, and osteoporosis rather than HDL-c in Chinese postmenopausal women with T2DM.
Collapse
|
38
|
Sabini E, Arboit L, Khan MP, Lanzolla G, Schipani E. Oxidative phosphorylation in bone cells. Bone Rep 2023; 18:101688. [PMID: 37275785 PMCID: PMC10238578 DOI: 10.1016/j.bonr.2023.101688] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
The role of energy metabolism in bone cells is an active field of investigation. Bone cells are metabolically very active and require high levels of energy in the form of adenosine triphosphate (ATP) to support their function. ATP is generated in the cytosol via glycolysis coupled with lactic acid fermentation and in the mitochondria via oxidative phosphorylation (OXPHOS). OXPHOS is the final convergent metabolic pathway for all oxidative steps of dietary nutrients catabolism. The formation of ATP is driven by an electrochemical gradient that forms across the mitochondrial inner membrane through to the activity of the electron transport chain (ETC) complexes and requires the presence of oxygen as the final electron acceptor. The current literature supports a model in which glycolysis is the main source of energy in undifferentiated mesenchymal progenitors and terminally differentiated osteoblasts, whereas OXPHOS appears relevant in an intermediate stage of differentiation of those cells. Conversely, osteoclasts progressively increase OXPHOS during differentiation until they become multinucleated and mitochondrial-rich terminal differentiated cells. Despite the abundance of mitochondria, mature osteoclasts are considered ATP-depleted, and the availability of ATP is a critical factor that regulates the low survival capacity of these cells, which rapidly undergo death by apoptosis. In addition to ATP, bioenergetic metabolism generates reactive oxygen species (ROS) and intermediate metabolites that regulate a variety of cellular functions, including epigenetics changes of genomic DNA and histones. This review will briefly discuss the role of OXPHOS and the cross-talks OXPHOS-glycolysis in the differentiation process of bone cells.
Collapse
Affiliation(s)
- Elena Sabini
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Lorenzo Arboit
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Giulia Lanzolla
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| |
Collapse
|
39
|
Yang Z, Zhang X, Zhuo F, Liu T, Luo Q, Zheng Y, Li L, Yang H, Zhang Y, Wang Y, Liu D, Tu P, Zeng K. Allosteric Activation of Transglutaminase 2 via Inducing an "Open" Conformation for Osteoblast Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206533. [PMID: 37088726 PMCID: PMC10288273 DOI: 10.1002/advs.202206533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/04/2023] [Indexed: 05/03/2023]
Abstract
Osteoblasts play an important role in the regulation of bone homeostasis throughout life. Thus, the damage of osteoblasts can lead to serious skeletal diseases, highlighting the urgent need for novel pharmacological targets. This study introduces chemical genetics strategy by using small molecule forskolin (FSK) as a probe to explore the druggable targets for osteoporosis. Here, this work reveals that transglutaminase 2 (TGM2) served as a major cellular target of FSK to obviously induce osteoblast differentiation. Then, this work identifies a previously undisclosed allosteric site in the catalytic core of TGM2. In particular, FSK formed multiple hydrogen bonds in a saddle-like domain to induce an "open" conformation of the β-sandwich domain in TGM2, thereby promoting the substrate protein crosslinks by incorporating polyamine. Furthermore, this work finds that TGM2 interacted with several mitochondrial homeostasis-associated proteins to improve mitochondrial dynamics and ATP production for osteoblast differentiation. Finally, this work observes that FSK effectively ameliorated osteoporosis in the ovariectomy mice model. Taken together, these findings show a previously undescribed pharmacological allosteric site on TGM2 for osteoporosis treatment, and also provide an available chemical tool for interrogating TGM2 biology and developing bone anabolic agent.
Collapse
Affiliation(s)
- Zhuo Yang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Xiao‐Wen Zhang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Fang‐Fang Zhuo
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Ting‐Ting Liu
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Qian‐Wei Luo
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Yong‐Zhe Zheng
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Ling Li
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Heng Yang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Yi‐Chi Zhang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Yan‐Hang Wang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Dan Liu
- Proteomics LaboratoryMedical and Healthy Analytical CenterPeking University Health Science CenterBeijing100191China
| | - Peng‐Fei Tu
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Ke‐Wu Zeng
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| |
Collapse
|
40
|
Li D, Gao Z, Li Q, Liu X, Liu H. Cuproptosis-a potential target for the treatment of osteoporosis. Front Endocrinol (Lausanne) 2023; 14:1135181. [PMID: 37214253 PMCID: PMC10196240 DOI: 10.3389/fendo.2023.1135181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Osteoporosis is an age-related disease of bone metabolism marked by reduced bone mineral density and impaired bone strength. The disease causes the bones to weaken and break more easily. Osteoclasts participate in bone resorption more than osteoblasts participate in bone formation, disrupting bone homeostasis and leading to osteoporosis. Currently, drug therapy for osteoporosis includes calcium supplements, vitamin D, parathyroid hormone, estrogen, calcitonin, bisphosphates, and other medications. These medications are effective in treating osteoporosis but have side effects. Copper is a necessary trace element in the human body, and studies have shown that it links to the development of osteoporosis. Cuproptosis is a recently proposed new type of cell death. Copper-induced cell death regulates by lipoylated components mediated via mitochondrial ferredoxin 1; that is, copper binds directly to the lipoylated components of the tricarboxylic acid cycle, resulting in lipoylated protein accumulation and subsequent loss of iron-sulfur cluster proteins, leading to proteotoxic stress and eventually cell death. Therapeutic options for tumor disorders include targeting the intracellular toxicity of copper and cuproptosis. The hypoxic environment in bone and the metabolic pathway of glycolysis to provide energy in cells can inhibit cuproptosis, which may promote the survival and proliferation of various cells, including osteoblasts, osteoclasts, effector T cells, and macrophages, thereby mediating the osteoporosis process. As a result, our group tried to explain the relationship between the role of cuproptosis and its essential regulatory genes, as well as the pathological mechanism of osteoporosis and its effects on various cells. This study intends to investigate a new treatment approach for the clinical treatment of osteoporosis that is beneficial to the treatment of osteoporosis.
Collapse
Affiliation(s)
- Dinglin Li
- Department of Integrated Traditional Chinese and Western Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhonghua Gao
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Li
- Department of Integrated Traditional Chinese and Western Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Liu
- Department of Integrated Traditional Chinese and Western Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Lopez N, Cohen SM, Emanuele M. Type 2 Diabetes and Bone Disease. Clin Rev Bone Miner Metab 2023; 21:21-31. [DOI: 10.1007/s12018-023-09288-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/10/2023] [Indexed: 01/05/2025]
|
42
|
Rashid S, Wilson SG, Zhu K, Walsh JP, Xu J, Mullin BH. Identification of Differentially Expressed Genes and Molecular Pathways Involved in Osteoclastogenesis Using RNA-seq. Genes (Basel) 2023; 14:genes14040916. [PMID: 37107674 PMCID: PMC10137460 DOI: 10.3390/genes14040916] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Osteoporosis is a disease that is characterised by reduced bone mineral density (BMD) and can be exacerbated by the excessive bone resorption of osteoclasts (OCs). Bioinformatic methods, including functional enrichment and network analysis, can provide information about the underlying molecular mechanisms that participate in the progression of osteoporosis. In this study, we harvested human OC-like cells differentiated in culture and their precursor peripheral blood mononuclear cells (PBMCs) and characterised the transcriptome of the two cell types using RNA-sequencing in order to identify differentially expressed genes. Differential gene expression analysis was performed in RStudio using the edgeR package. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to identify enriched GO terms and signalling pathways, with inter-connected regions characterised using protein-protein interaction analysis. In this study, we identified 3201 differentially expressed genes using a 5% false discovery rate; 1834 genes were upregulated, whereas 1367 genes were downregulated. We confirmed a significant upregulation of several well-established OC genes including CTSK, DCSTAMP, ACP5, MMP9, ITGB3, and ATP6V0D2. The GO analysis suggested that upregulated genes are involved in cell division, cell migration, and cell adhesion, while the KEGG pathway analysis highlighted oxidative phosphorylation, glycolysis and gluconeogenesis, lysosome, and focal adhesion pathways. This study provides new information about changes in gene expression and highlights key biological pathways involved in osteoclastogenesis.
Collapse
Affiliation(s)
- Sarah Rashid
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6907, Australia
| | - Scott G Wilson
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6907, Australia
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Kun Zhu
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
- Medical School, University of Western Australia, Perth, WA 6907, Australia
| | - John P Walsh
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
- Medical School, University of Western Australia, Perth, WA 6907, Australia
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6907, Australia
| | - Benjamin H Mullin
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6907, Australia
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| |
Collapse
|
43
|
Xie D, Xu Y, Zhang Y, Cai W, Lan X, Yan H. Pyruvate dehydrogenase kinase 1–dependent metabolic reprogramming: A promising target for postmenopausal osteoporosis treatment. Biomed Pharmacother 2023. [DOI: 10.1016/j.biopha.2023.114411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023] Open
|
44
|
Roberts FL, MacRae VE. Bone mineralisation and glucose metabolism. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2023; 29:100446. [PMID: 39184263 PMCID: PMC11339533 DOI: 10.1016/j.coemr.2023.100446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/30/2023] [Accepted: 03/03/2023] [Indexed: 08/27/2024]
Abstract
Recent advancements in the bone biology field have identified a novel bone-metabolism axis. In this review, we highlight several novel studies that further our knowledge of new endocrine functions of bone; explore remaining unanswered questions; and discuss translational challenges in this complex era of bone biology research.
Collapse
Affiliation(s)
| | - Vicky E. MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| |
Collapse
|
45
|
Deng X, Kato H, Taguchi Y, Nakata T, Umeda M. Intracellular glucose starvation inhibits osteogenic differentiation in human periodontal ligament cells. J Periodontal Res 2023; 58:607-620. [PMID: 36883427 DOI: 10.1111/jre.13112] [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: 07/05/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Periodontal ligament cells (PDLCs), as mesenchymal cells in the oral cavity, are closely linked to periodontal tissue regeneration. However, the effect of local glucose deficiency on periodontal tissue regeneration, such as immediately post-surgery, remains unknown. OBJECTIVE In the present study, we investigated the effect of a low-glucose environment on the proliferation and osteogenic differentiation of PDLCs. MATERIALS AND METHODS We used media with five glucose concentrations (100, 75, 50, 25, and 0 mg/dL) and focused on the effects of a low-glucose environment on the proliferation, osteogenic differentiation, and autophagy of PDLCs. Additionally, we focused on changes in lactate production in a low-glucose environment and investigated the involvement of lactate with AZD3965, a monocarboxylate transporter-1 (MCT-1) inhibitor. RESULTS The low-glucose environment inhibited PDLCs proliferation, migration, and osteogenic differentiation, and induced the expression of the autophagy-related factors LC3 and p62. Lactate and ATP production were decreased under low-glucose conditions. The addition of AZD3965 (MCT-1 inhibitor) in normal glucose conditions caused a similar trend as in low-glucose conditions on PDLCs. CONCLUSION Our results suggest lactate production through glucose metabolism in the osteogenic differentiation of PDLCs. A low-glucose environment decreased lactate production, inhibiting cell proliferation, migration, and osteogenic differentiation and inducing autophagy in PDLCs.
Collapse
Affiliation(s)
- Xin Deng
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Hirohito Kato
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Yoichiro Taguchi
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Takaya Nakata
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Makoto Umeda
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
46
|
Lungu O, Toscani D, Burroughs-Garcia J, Giuliani N. The Metabolic Features of Osteoblasts: Implications for Multiple Myeloma (MM) Bone Disease. Int J Mol Sci 2023; 24:ijms24054893. [PMID: 36902326 PMCID: PMC10003241 DOI: 10.3390/ijms24054893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The study of osteoblast (OB) metabolism has recently received increased attention due to the considerable amount of energy used during the bone remodeling process. In addition to glucose, the main nutrient for the osteoblast lineages, recent data highlight the importance of amino acid and fatty acid metabolism in providing the fuel necessary for the proper functioning of OBs. Among the amino acids, it has been reported that OBs are largely dependent on glutamine (Gln) for their differentiation and activity. In this review, we describe the main metabolic pathways governing OBs' fate and functions, both in physiological and pathological malignant conditions. In particular, we focus on multiple myeloma (MM) bone disease, which is characterized by a severe imbalance in OB differentiation due to the presence of malignant plasma cells into the bone microenvironment. Here, we describe the most important metabolic alterations involved in the inhibition of OB formation and activity in MM patients.
Collapse
Affiliation(s)
- Oxana Lungu
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Hematology, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy
- Correspondence:
| |
Collapse
|
47
|
Wang Z, Zhang X, Cheng X, Ren T, Xu W, Li J, Wang H, Zhang J. Inflammation produced by senescent osteocytes mediates age-related bone loss. Front Immunol 2023; 14:1114006. [PMID: 36814916 PMCID: PMC9940315 DOI: 10.3389/fimmu.2023.1114006] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/11/2023] [Indexed: 02/08/2023] Open
Abstract
Purpose The molecular mechanisms of age-related bone loss are unclear and without valid drugs yet. The aims of this study were to explore the molecular changes that occur in bone tissue during age-related bone loss, to further clarify the changes in function, and to predict potential therapeutic drugs. Methods We collected bone tissues from children, middle-aged individuals, and elderly people for protein sequencing and compared the three groups of proteins pairwise, and the differentially expressed proteins (DEPs) in each group were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). K-means cluster analysis was then used to screen out proteins that continuously increased/decreased with age. Canonical signaling pathways that were activated or inhibited in bone tissue along with increasing age were identified by Ingenuity Pathway Analysis (IPA). Prediction of potential drugs was performed using the Connectivity Map (CMap). Finally, DEPs from sequencing were verified by Western blot, and the drug treatment effect was verified by quantitative real-time PCR. Results The GO and KEGG analyses show that the DEPs were associated with inflammation and bone formation with aging, and the IPA analysis shows that pathways such as IL-8 signaling and acute-phase response signaling were activated, while glycolysis I and EIF2 signaling were inhibited. A total of nine potential drugs were predicted, with rapamycin ranking the highest. In cellular experiments, rapamycin reduced the senescence phenotype produced by the H2O2-stimulated osteocyte-like cell MLO-Y4. Conclusion With age, inflammatory pathways are activated in bone tissue, and signals that promote bone formation are inhibited. This study contributes to the understanding of the molecular changes that occur in bone tissue during age-related bone loss and provides evidence that rapamycin is a drug of potential clinical value for this disease. The therapeutic effects of the drug are to be further studied in animals.
Collapse
Affiliation(s)
- Zixuan Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Zhang
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Cheng
- Health Care Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianxing Ren
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weihua Xu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Wang
- Department of Medical Genetics, Basic School of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Jinxiang Zhang, ; Hui Wang,
| | - Jinxiang Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Jinxiang Zhang, ; Hui Wang,
| |
Collapse
|
48
|
Shen YW, Cheng YA, Li Y, Li Z, Yang BY, Li X. Sambucus williamsii Hance maintains bone homeostasis in hyperglycemia-induced osteopenia by reversing oxidative stress via cGMP/PKG signal transduction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154607. [PMID: 36610352 DOI: 10.1016/j.phymed.2022.154607] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 11/30/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Sambucus williamsii Hance (SWH) has effectively been adopted to treat joint and bone disorders. Diabetes-induced osteopenia (DOP) is caused primarily by impaired bone formation as a result of hyperglycemia. We had previously demonstrated that SWH extract accelerated fracture healing and promoted osteoblastic MC3T3-E1 cell proliferation and osteogenic differentiation. This study assessed the impacts of SWH extract on diabetes-induced bone loss and explored the mechanisms underlying its osteoprotective effects. METHODS This work employed MC3T3-E1 cell line for evaluating how SWH extract affected osteogenesis, oxidative stress (OS), and the underlying mechanism in vitro. Streptozotocin-induced osteopenia mouse model was applied with the purpose of assessing SWH extract's osteoprotection on bone homeostasis in vivo. RESULTS The increased OS of MC3T3-E1 cells exposed to high glucose (HG) was largely because of the upregulation of pro-oxidant genes and the downregulation of antioxidant genes, whereas SWH extract reduced the OS by modulating NADPH oxidase-4 and thioredoxin-related genes by activating cyclic guanosine monophosphate (cGMP) production and increasing the level of cGMP-mediated protein kinase G type-2 (PKG2). The oral administration of SWH extract maintained bone homeostasis in type 1 diabetes mellitus (T1DM) mice by enhancing osteogenesis while decreasing OS. In bones from hyperglycemia-induced osteopenia mice and HG-treated MC3T3-E1 cells, the SWH extract achieved the osteoprotective effects through activating the cGMP/PKG2 signaling pathway, upregulating the level of antioxidant genes, as well as downregulating the level of pro-oxidant genes. CONCLUSION SWH extract exerts osteoprotective effects on hyperglycemia-induced osteopenia by reversing OS via cGMP/PKG signal transduction and is a potential therapy for DOP.
Collapse
Affiliation(s)
- Yi-Wei Shen
- Ningbo Hospital of Traditional Chinese Medicine (Ningbo Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medicine University), Ningbo, Zhejiang, 315010, China; The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, Heilongjiang 150040, China; Key Laboratory of Northern Medicine Base and Application under Ministry of d Education, Harbin, Heilongjiang 150040, China; Key Laboratory of Chinese Materia Medica, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Yang-Ang Cheng
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, Heilongjiang 150040, China; Key Laboratory of Northern Medicine Base and Application under Ministry of d Education, Harbin, Heilongjiang 150040, China
| | - Yi Li
- College of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Zuo Li
- College of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Bing-You Yang
- College of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Xue Li
- Ningbo Hospital of Traditional Chinese Medicine (Ningbo Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medicine University), Ningbo, Zhejiang, 315010, China; The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, Heilongjiang 150040, China.
| |
Collapse
|
49
|
Bow AJ, Rifkin RE, Priester C, Christopher CJ, Grzeskowiak RM, Hecht S, Adair SH, Mulon PY, Castro HF, Campagna SR, Anderson DE. Temporal metabolic profiling of bone healing in a caprine tibia segmental defect model. Front Vet Sci 2023; 9:1023650. [PMID: 36733424 PMCID: PMC9886884 DOI: 10.3389/fvets.2022.1023650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
Bone tissue engineering is an emerging field of regenerative medicine, with a wide array of biomaterial technologies and therapeutics employed. However, it is difficult to objectively compare these various treatments during various stages of tissue response. Metabolomics is rapidly emerging as a powerful analytical tool to establish broad-spectrum metabolic signatures for a target biological system. Developing an effective biomarker panel for bone repair from small molecule data would provide an objective metric to readily assess the efficacy of novel therapeutics in relation to natural healing mechanisms. In this study we utilized a large segmental bone defect in goats to reflect trauma resulting in substantial volumetric bone loss. Characterization of the native repair capacity was then conducted over a period of 12 months through the combination of standard (radiography, computed tomography, histology, biomechanics) data and ultra-high-performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS) metabolic profiling. Standard metrics demonstrated that samples formed soft callus structures that later mineralized. Small molecule profiles showed distinct temporal patterns associated with the bone tissue repair process. Specifically, increased lactate and amino acid levels at early time points indicated an environment conducive to osteoblast differentiation and extracellular matrix formation. Citrate and pyruvate abundances increased at later time points indicating increasing mineral content within the defect region. Taurine, shikimate, and pantothenate distribution profiles appeared to represent a shift toward a more homeostatic remodeling environment with the differentiation and activity of osteoclasts offsetting the earlier deposition phases of bone repair. The generation of a comprehensive metabolic reference portfolio offers a potent mechanism for examining novel biomaterials and can serve as guide for the development of new targeted therapeutics to improve the rate, magnitude, and quality of bone regeneration.
Collapse
Affiliation(s)
- Austin J. Bow
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States,*Correspondence: Austin J. Bow ✉
| | - Rebecca E. Rifkin
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, Knoxville, TN, United States
| | | | - Remigiusz M. Grzeskowiak
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Silke Hecht
- Department of Small Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Steve H. Adair
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Pierre-Yves Mulon
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Hector F. Castro
- Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States,Biological and Small Molecule Mass Spectrometry Core and the Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Shawn R. Campagna
- Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States,Biological and Small Molecule Mass Spectrometry Core and the Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - David E. Anderson
- University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States,David E. Anderson ✉
| |
Collapse
|
50
|
Fraser D, Ganesan SM. Microbiome, alveolar bone, and metabolites: Connecting the dots. FRONTIERS IN DENTAL MEDICINE 2023. [DOI: 10.3389/fdmed.2022.1074339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The oral microbiome (OM) is a diverse and dynamic collection of species, separated from alveolar bone by the oral mucosa. Pathogenic shifts in the OM (dysbiosis) during periodontitis are associated with an inflammatory response in the oral mucosa that drives alveolar bone resorption. Alveolar bone is also affected by metabolic disorders such as osteoporosis. Accumulating evidence has linked another microbial community, the gut microbiome (GM), to systemic bone metabolism and osteoporosis. Underlying this connection is the biologic activity of metabolites, byproducts of host and bacterial activity. Limited evidence also suggests that metabolites in the oral cavity signal between the OM and immune system, influencing both alveolar bone homeostasis and pathologic bone destruction in periodontitis. While the oral cavity and gut are connected through the gastrointestinal tract, dissimilar roles for known metabolites between these two niches exemplify the difficulty in translating knowledge on gut-derived metabolites and bone metabolism to alveolar bone. Integrated metabolomic, transcriptomic, and metagenomic approaches hold promise for resolving these challenges and identifying novel metabolites which impact alveolar bone health. Further interrogation through mechanistic testing in pre-clinical models and carefully controlled clinical studies have potential to lead toward translation of these discoveries into meaningful therapies.
Collapse
|