1
|
Tu Y, Miao J, Wu Q, Lu K, Ren R, Lin C, Wang X, Jin H. Obstructive sleep apnea and osteoporosis: A bidirectional two-sample mendelian randomization analysis. Respir Med 2025; 242:108090. [PMID: 40215796 DOI: 10.1016/j.rmed.2025.108090] [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: 02/29/2024] [Revised: 11/28/2024] [Accepted: 04/04/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND Previous epidemiological studies have explored the association between obstructive sleep apnea (OSA) and osteoporosis (OP), with inconclusive results due to various biases. Herein, we sought to determine the causal association between OSA and OP through bidirectional two-sample Mendelian randomization analysis. METHODS Summary-level data for OSA were acquired from the FinnGen consortium, while data for fractures and BMDs (FA-BMD, FN-BMD, LS-BMD and eBMD) were derived from the UKBB and GEFOS. The inverse variance weighted (IVW) method was conducted as the main method, and several supplementary methods were further utilized for sensitivity analysis to strengthen the reliability of our findings. RESULTS The study findings strongly suggest a causal association between OSA and FA-BMD based on the IVW method (BETA = 0.404; 95 % CI = 0.208, 0.599; p = 5.28 × 10-5). However, OSA showed no significant causal relationship with eBMD (BETA = 0.052; 95 % CI = -0.018, 0.123; p = 0.145), FN-BMD (BETA = 0.095; 95 % CI = -0.009, 0.2; p = 0.073), LS-BMD (BETA = 0.021; 95 % CI = -0.082, 0.124; p = 0.695), and fractures (OR = 0.998; 95 % CI = 0.907, 1.098; p = 0.971). The conclusions from other analytical strategies were generally aligned with those of the IVW. No definitive causal effect of OP on OSA was observed in reverse analysis. CONCLUSION This research provided clear evidence of a causal association between OSA and FA-BMD, shedding light on the potential impact of OSA on bone metabolism.
Collapse
Affiliation(s)
- Yiting Tu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, China.
| | - Jiansen Miao
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, China.
| | - Qihang Wu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, China.
| | - Keyu Lu
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, China.
| | - Rufeng Ren
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Chihao Lin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, China.
| | - Xiangyang Wang
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, China.
| | - Haiming Jin
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China; Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, China.
| |
Collapse
|
2
|
Watts LM, Sparkes PC, Dewhurst HF, Guilfoyle SE, Pollard AS, Komla-Ebri D, Butterfield NC, Williams GR, Bassett JHD. The GWAS candidate far upstream element binding protein 3 (FUBP3) is required for normal skeletal growth, and adult bone mass and strength in mice. Bone 2025; 195:117472. [PMID: 40139337 DOI: 10.1016/j.bone.2025.117472] [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: 12/31/2024] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Bone mineral density (BMD) and height are highly heritable traits for which hundreds of genetic loci have been linked through genome wide association studies (GWAS). FUBP3 is a DNA and RNA binding protein best characterised as a transcriptional regulator of c-Myc, but little is known about its role in vivo. Single nucleotide polymorphisms in FUBP3 at the 9q34.11 locus have been associated with BMD, fracture and height in multiple GWAS, but FUBP3 has no previously established role in the skeleton. We analysed Fubp3-deficient mice to determine the consequence of FUBP3 deficiency in vivo. Mice lacking Fubp3 had reduced survival to adulthood and impaired skeletal growth. Bone mass was decreased, most strikingly in the vertebrae, with altered trabecular micro-architecture. Fubp3 deficient bones were also weak. These data provide the first functional demonstration that Fubp3 is required for normal skeletal growth and development and maintenance of adult bone structure and strength, indicating that FUBP3 contributes to the GWAS association of 9q34.11 with variation in height, BMD and fracture.
Collapse
Affiliation(s)
- Laura M Watts
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Penny C Sparkes
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Hannah F Dewhurst
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Siobhan E Guilfoyle
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Andrea S Pollard
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Davide Komla-Ebri
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Natalie C Butterfield
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
3
|
Cai J, Huang H, Hu H, Qi L, Zhou T. Association of nuclear magnetic resonance-based metabolomics with bone health in the UK Biobank. Bone 2025; 195:117460. [PMID: 40118262 DOI: 10.1016/j.bone.2025.117460] [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: 12/03/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVES The study aimed to explore associations of metabolomic data based on nuclear magnetic resonance (NMR) with the risk of fractures and bone mineral density (BMD). METHODS We included 69,963 participants without fractures at baseline in the UK Biobank. Cox proportional hazard models were used to estimate the associations of metabolomic biomarkers measured by NMR technology with the risk of all fractures and hip fracture. We used principal component analysis (PCA) to obtain uncorrelated principal components (PC), which were further used to estimate the associations of each PC with BMD, all fractures, and hip fracture separately. RESULTS During a median follow-up of 12.6 years, 3840 incidents of all fractures and 666 incidents of hip fracture were documented. Ninety-four of the 143 metabolomic biomarkers were significantly associated with incident all fractures, and 81 were significantly associated with incident hip fracture. The very low-density lipoprotein (VLDL) subclasses in different lipid constituents were associated with increased BMD at multiple sites, whereas high-density lipoprotein (HDL) subclasses were associated with decreased BMD. Higher concentrations of small (HR per SD increment: 0.92; 95 % CI: 0.88-0.97), medium (HR per SD increment: 0.91; 95 % CI: 0.88-0.94), and large (HR per SD increment: 0.93; 95 % CI: 0.90-0.96) low-density lipoprotein (LDL) particles were associated with a lower risk of all fractures. Similarly, higher VLDL subclasses (excluding very small VLDL particles) were associated with a lower risk of all fractures. Besides, higher levels of lipid constituents (including total lipids, cholesteryl esters, cholesterol, and free cholesterol) of very large and large HDL were associated with an increased risk of all fractures. PC1 (mainly contributed by lipid subclasses of LDL and VLDL), which explained the most variance of individual biomarkers, showed a negative association with the risk of all fractures (P = 7.80E-08). Similar associations were observed for hip fracture. CONCLUSIONS Higher levels of large and very large HDL were associated with an increased risk of fractures, whereas higher lipid subclasses of LDL and VLDL were associated with a lower risk of fracture. Higher levels of VLDL subclasses in different lipid constituents were associated with increased BMD at multiple sites, while higher level of HDL was associated with decreased BMD.
Collapse
Affiliation(s)
- Jie Cai
- Department of Epidemiology, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Huan Huang
- Department of Epidemiology, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Huaying Hu
- Department of Epidemiology, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States of America; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America.
| | - Tao Zhou
- Department of Epidemiology, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
4
|
Greene CS, Gignoux CR, Subirana-Granés M, Pividori M, Hicks SC, Ackert-Bicknell CL. Can AI reveal the next generation of high-impact bone genomics targets? Bone Rep 2025; 25:101839. [PMID: 40225702 PMCID: PMC11986539 DOI: 10.1016/j.bonr.2025.101839] [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: 01/08/2025] [Revised: 03/18/2025] [Accepted: 03/23/2025] [Indexed: 04/15/2025] Open
Abstract
Genetic studies have revealed hundreds of loci associated with bone-related phenotypes, including bone mineral density (BMD) and fracture risk. However, translating discovered loci into effective new therapies remains challenging. We review success stories including PCSK9-related drugs in cardiovascular disease and evidence supporting the use of human genetics to guide drug discovery, while highlighting advances in artificial intelligence and machine learning with the potential to improve target discovery in skeletal biology. These strategies are poised to improve how we integrate diverse data types, from genetic and electronic health records data to single-cell profiles and knowledge graphs. Such emerging computational methods can position bone genomics for a future of more precise, effective treatments, ultimately improving the outcomes for patients with common and rare skeletal disorders.
Collapse
Affiliation(s)
- Casey S. Greene
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christopher R. Gignoux
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marc Subirana-Granés
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Milton Pividori
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Cheryl L. Ackert-Bicknell
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
5
|
Qiu H, Jin H, Miao J, Li H, Chen J, Yang X, Chen X, Mullin BH, Chen K, Gu R, Qin A, Wilson SG, Xu J. Heme metabolism mediates RANKL-induced osteoclastogenesis via mitochondrial oxidative phosphorylation. J Bone Miner Res 2025; 40:639-655. [PMID: 40073838 PMCID: PMC12103724 DOI: 10.1093/jbmr/zjaf040] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Bone undergoes life-long remodeling, in which disorders of bone remodeling could occur in many pathological conditions including osteoporosis. Understanding the cellular metabolism of osteoclasts (OCs) is key to developing new treatments for osteoporosis, a disease that affects over 200 million women worldwide per annum. We found that human OC differentiation from peripheral blood mononuclear cells derived from 8 female patients is featured with a distinct gene expression profile of mitochondrial biogenesis. Elevated mitochondrial membrane potential (MMP, Δψm) was also observed in receptor activator of NF-κB ligand (RANKL)-induced OCs. Interestingly, the gene pathways of heme synthesis and metabolism were activated upon RANKL stimulation, featured by transcriptomic profiling in murine cells at a single-cell resolution, which revealed a stepwise expression pattern of heme-related genes. The real-world human data also divulges potential links between heme-related genes and bone mineral density. Heme is known to have a role in the formation of functional mitochondrial complexes that regulate MMP. Disruption of heme biosynthesis via genetically silencing Ferrochelatase or a selective inhibitor, N-methyl Protoporphyrin IX (NMPP), demonstrated potent inhibition of OC differentiation, with a dose-dependent effect observed in NMPP treatment and a substantial efficacy even at a single dose. In vivo study further showed the protective effect of NMPP on ovariectomy-induced bone loss in female mice. Collectively, we found that RANKL-mediated signaling regulated mitochondrial formation and heme metabolism to synergistically support osteoclastogenesis. Inhibition of heme synthesis impaired OC formation and reversed excessive bone loss, representing a new therapeutic target for metabolic skeletal disorders.
Collapse
Affiliation(s)
- Heng Qiu
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, 6009, Australia
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Haiming Jin
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, 6009, Australia
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiansen Miao
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Hui Li
- Shenzhen University of Advanced Technology, and Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Junchun Chen
- Shenzhen University of Advanced Technology, and Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiaohong Yang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510220, China
| | - Xiaojun Chen
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Benjamin H Mullin
- Department of Endocrinology & Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
- Department of Endocrinology & Diabetes, Medical School, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Kai Chen
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, 6009, Australia
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Ronghe Gu
- Department of Orthopedics, First People's Hospital of Nanning, Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, China
| | - An Qin
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, 6009, Australia
- Shanghai Key Laboratory of Orthopaedic Implant, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Scott G Wilson
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, 6009, 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, WC2R 2LS, United Kingdom
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, 6009, Australia
- Shenzhen University of Advanced Technology, and Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
6
|
Sha L, Zhang L, Zhao X, Xiang R, Wu X, Zhu J, Hou J, Deng Q, Qin C, Xiao C, Qu Y, Han T, Zhou J, Zheng S, Yu T, Song X, Yang B, Fan M, Jiang X. Shared Genetic Architecture and Causal Relationship Between Serum 25-Hydroxyvitamin D and Bone Mineral Density. J Clin Endocrinol Metab 2025; 110:1605-1616. [PMID: 39431290 DOI: 10.1210/clinem/dgae738] [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: 07/13/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024]
Abstract
CONTEXT Despite the well-established regulatory role of vitamin D in maintaining bone health, little is known about the shared genetics and causality of the association between serum 25-hydroxyvitamin D (25OHD) and bone mineral density (BMD). OBJECTIVE We aimed to investigate the shared genetic architecture and causal relationship between serum 25OHD and BMD, providing insights into their underlying biological mechanisms. METHODS Leveraging individual-level data from the UK Biobank (UKB) cohort and summary-level data from the genome-wide association studies (GWASs) conducted on European individuals for serum 25OHD (N = 417 580) and estimated heel BMD (eBMD, N = 426 824), we systematically elucidated the shared genetic architecture underlying serum 25OHD and eBMD through a comprehensive genome-wide cross-trait design. RESULTS Despite a lack of global genetic correlation (rg=-0.001; P = .95), a statistically significant local signal was discovered at 5p11-5q11.9. Two-sample mendelian randomization (MR) indicated no causal association in the overall population (β=.003, 95% CI, -0.04 to 0.03; P = .93), while positive causal effects were observed in males (β=.005, 95% CI, 0.00 to 0.01; P = .03) and older individuals (β=.009, 95% CI, 0.00∼0.02; P = .01) according to one-sample MR. A total of 49 pleiotropic single-nucleotide variations (SNVs), with 4 novel SNVs (rs1077151, rs79873740, rs12150353, and rs4760401), were identified, and a total of 95 gene-tissue pairs exhibited overlap, predominantly enriched in the nervous, digestive, exocrine/endocrine, and cardiovascular systems. Protein-protein interaction analysis identified RPS9 and RPL7A as hub genes. CONCLUSION This study illuminates the potential health benefits of enhancing serum 25OHD levels to mitigate the risk of osteoporosis among men and individuals older than 65 years. It also unveils a shared genetic basis between serum 25OHD and eBMD, offering valuable insights into the intricate biological pathways.
Collapse
Affiliation(s)
- Linna Sha
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Li Zhang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rong Xiang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiangbo Zhu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiaojiao Hou
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qin Deng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chenjiarui Qin
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Changfeng Xiao
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yang Qu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Tao Han
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jinyu Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Sirui Zheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ting Yu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xin Song
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Bin Yang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mengyu Fan
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xia Jiang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| |
Collapse
|
7
|
Duan Y, Zhao LJ, Lu YT, Li J, Li SX. Crosstalk between kidney and bones: New perspective for modulating osteoporosis. Ageing Res Rev 2025; 109:102776. [PMID: 40389172 DOI: 10.1016/j.arr.2025.102776] [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: 12/27/2024] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Growing evidence indicates an interesting interplay between kidney and bone. The pathophysiological condition of the skeletal system is intricately associated with the normal functioning of the kidneys. This relationship is modulated by various factors, including calcium and phosphate, 1-α-hydroxylase, erythropoietin (EPO), klotho, fibroblast growth factor 23 (FGF23), bone morphogenetic protein-7 (BMP-7), and extracellular vesicles (EVs). These interactions are notably evident in conditions such as chronic kidney disease with bone mineral density (CKD-BMD), renal osteodystrophy (ROD), and osteoporosis (OP). Furthermore, innovative methodologies such as cell co-culture, organ-on-a-chip, single-cell sequencing, and spatial transcriptomics are highlighted as instrumental in advancing the study of inter-organ interactions. This review, grounded in the pathogenesis, diagnostic and therapeutic modalities, and pharmacological treatments of OP, synthesizes evidence from molecular biology to clinical perspectives. It aims to establish a foundation for the development of more complex and physiologically relevant in vitro models and to propose potential therapeutic strategies.
Collapse
Affiliation(s)
- Yan Duan
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Engineering Technology Research Center for Bioactive Substance Discovery of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Province Sino-US International Joint Research Center for Therapeutic Drugs of Senile Degenerative Diseases, Changsha, Hunan 410208, PR China
| | - Li-Juan Zhao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Engineering Technology Research Center for Bioactive Substance Discovery of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Province Sino-US International Joint Research Center for Therapeutic Drugs of Senile Degenerative Diseases, Changsha, Hunan 410208, PR China; College of Biology and Food Engineering, Huai Hua University, Huaihua 418000, PR China
| | - Yu-Ting Lu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Engineering Technology Research Center for Bioactive Substance Discovery of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Province Sino-US International Joint Research Center for Therapeutic Drugs of Senile Degenerative Diseases, Changsha, Hunan 410208, PR China; Department of Medicine, Guangxi University of Science and Technology, Liuzhou 545005, PR China
| | - Juan Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Engineering Technology Research Center for Bioactive Substance Discovery of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Province Sino-US International Joint Research Center for Therapeutic Drugs of Senile Degenerative Diseases, Changsha, Hunan 410208, PR China.
| | - Shun-Xiang Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Engineering Technology Research Center for Bioactive Substance Discovery of Chinese Medicine, Changsha, Hunan 410208, PR China; Hunan Province Sino-US International Joint Research Center for Therapeutic Drugs of Senile Degenerative Diseases, Changsha, Hunan 410208, PR China.
| |
Collapse
|
8
|
Wu Q, Jung J. Precision fracture risk assessment: leveraging genomic and clinical data for personalized care. Osteoporos Int 2025:10.1007/s00198-025-07517-x. [PMID: 40329100 DOI: 10.1007/s00198-025-07517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Affiliation(s)
- Qing Wu
- Department of Biomedical Informatics, College of Medicine, Center for Biostatistics (Dr. Qing Wu, Jongyun Jung), The Ohio State University Wexner Medical Center, 250 Lincoln Tower, 1800 Cannon Drive, Columbus, OH, USA.
| | - Jongyun Jung
- Department of Biomedical Informatics, College of Medicine, Center for Biostatistics (Dr. Qing Wu, Jongyun Jung), The Ohio State University Wexner Medical Center, 250 Lincoln Tower, 1800 Cannon Drive, Columbus, OH, USA
| |
Collapse
|
9
|
Wu Q, Jung J. Ensemble-learning approach improves fracture prediction using genomic and phenotypic data. Osteoporos Int 2025; 36:811-821. [PMID: 40053072 PMCID: PMC12089207 DOI: 10.1007/s00198-025-07437-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 02/14/2025] [Indexed: 05/20/2025]
Abstract
This study presents an innovative ensemble machine learning model integrating genomic and clinical data to enhance the prediction of major osteoporotic fractures in older men. The Super Learner (SL) model achieved superior performance (AUC = 0.76, accuracy = 95.6%, sensitivity = 94.5%, specificity = 96.1%) compared to individual models. Ensemble machine learning improves fracture prediction accuracy, demonstrating the potential for personalized osteoporosis management. PURPOSE Existing fracture risk models have limitations in their accuracy and in integrating genomic data. This study developed and validated an innovative ensemble machine learning (ML) model that combines multiple algorithms and integrates clinical, lifestyle, skeletal, and genomic data to enhance prediction for major osteoporotic fractures (MOF) in older men. METHODS This study analyzed data from 5130 participants in the Osteoporotic Fractures in Men cohort Study. The model incorporated 1103 individual genome-wide significant variants and conventional risk factors of MOF. The participants were randomly divided into training (80%) and testing (20%) sets. Seven ML algorithms were combined using the SL ensemble method with tenfold cross-validation MOF prediction. Model performance was evaluated on the testing set using the area under the curve (AUC), the area under the precision-recall curve, calibration, accuracy, sensitivity, specificity, negative predictive value (NPV), positive predictive value (PPV), and reclassification metrics. SL model performances were evaluated by comparison with baseline models and subgroup analyses by race. RESULTS The SL model demonstrated the best performance with an AUC of 0.76, accuracy of 95.6%, sensitivity of 94.5%, specificity of 96.1%, NPV of 95.1%, and PPV of 94.7%. Among the individual ML, gradient boosting performed optimally. The SL model outperformed baseline models, and it also achieved accuracies of 93.1% for Whites and 91.6% for Minorities, outperforming single ML in subgroup analysis. CONCLUSION The ensemble learning approach significantly improved fracture prediction accuracy and model performance compared to individual ML. Integrating genomic and phenotypic data via the SL approach represents a promising advancement for personalized osteoporosis management.
Collapse
Affiliation(s)
- Qing Wu
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, USA.
| | - Jongyun Jung
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, USA
| |
Collapse
|
10
|
Dong X, Liu H, Yuan D, Gulati K, Liu Y. Re-engineering bone: pathogenesis, diagnosis and emerging therapies for osteoporosis. J Mater Chem B 2025; 13:4938-4963. [PMID: 40192254 DOI: 10.1039/d4tb02628d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Osteoporosis, a multifaceted metabolic bone disease, is becoming increasingly prevalent and poses a significant burden on global healthcare systems. Given the limitations of traditional treatments such as pharmacotherapy, tissue engineering has emerged as a promising alternative for osteoporosis management. This review begins by exploring the pathogenesis of osteoporosis, with a focus on the abnormal metabolic, cellular, and molecular signalling microenvironments that drive the disease. We also examine commonly used clinical diagnostic techniques, discussing their strengths and limitations. Notably, this review evaluates various advanced tissue engineering strategies for osteoporosis treatment. Delivery systems, including injectable hydrogels and nanomaterials, are detailed alongside bone tissue engineering materials such as bioactive ceramics, bone cements, and polymers. Additionally, biologically active substances, including exosomes and cytokines, and emerging therapies that leverage small-molecule drugs are explored. Through a comprehensive analysis of the advantages and limitations of current biomaterials and therapeutic approaches, this review provides insights into future directions for tissue engineering-based solutions. By synthesizing current advancements, it aims to inspire innovative perspectives for the clinical management of osteoporosis.
Collapse
Affiliation(s)
- Xinyi Dong
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
- National Center for Stomatology & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology & Translational Research Center for Oro-craniofacial Stem Cells and Systemic Health, Beijing 100081, China
| | - Hao Liu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
- National Center for Stomatology & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices Beijing Key Laboratory of Digital Stomatology & Translational Research Center for Oro-craniofacial Stem Cells and Systemic Health, Beijing 100081, China
| | - Dian Yuan
- Hubei University of Science and Technology, School of Dentistry and Optometry, Xianning 430030, China
| | - Karan Gulati
- School of Dentistry, The University of Queensland, Herston, QLD, 4006, Australia.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), Herston, QLD 4006, Australia
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
11
|
Rudra S, Kennedy K, Neukrug S, Huang J, Cousminer DL, Patel A, Xu Y, Grant SFA, Baldassano RN, Albenberg L, Zemel BS, Stein R. Prevalence and predictors of low bone mineral density in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2025. [PMID: 40296588 DOI: 10.1002/jpn3.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVES Bone health is at risk in children with inflammatory bowel disease (IBD). This study examined the prevalence and predictors of low bone mineral density (BMD) in a cohort of children and young adults with IBD. METHODS This single-center retrospective study included patients with IBD, ages 3.5-22 years, with completed dual x-ray absorptiometry (DXA) scans from 2006 to 2019. Demographic, clinical, and laboratory data were collected. Logistic regression analysis identified predictors associated with low BMD (Z-scores ≤ -2 standard deviations [SDs]) for three outcomes. In an overlapping IBD cohort with available genetic data between 2002 and 2019 (n = 378), genetic risk for diminished bone health was calculated using published polygenic risk scores generated from genome-wide association studies based on DXA or heel ultrasound speed of sound (SOS). Linear regression analysis examined associations of low BMD and genetic risk. RESULTS Low BMD prevalence was 7% in our cohort (n = 600) based on spine bone mineral apparent density (BMAD), which best accounts for growth delays. Median (interquartile range [IQR]) spine BMAD Z-score was -0.37 SD (-1.11 to 0.35). Predictors of low BMAD included lower BMI Z-score (odds ratio [OR]: 0.67, p value: 0.02) and decreased height Z-score (OR: 0.6, p value: 0.005). Of those with longitudinal data (n = 118), low BMI (OR: 0.44, p value: <0.001) and steroid use (OR: 3.42, p value: 0.01) were associated with suboptimal bone health (Z-scores ≤ -1SD). In the cohort with genetic data, heel genomic SOS (β [standard error] = 0.17 [0.35], p ≤ 0.01) was associated with BMD. CONCLUSIONS Lower BMI should prompt DXA monitoring in pediatric IBD. Genetic predisposition may identify an at-risk subpopulation.
Collapse
Affiliation(s)
- Sharmistha Rudra
- Division of Gastroenterology, Hepatology, & Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, Georgia, USA
| | - Kanak Kennedy
- Division of Gastroenterology, Hepatology, & Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sarah Neukrug
- Division of Gastroenterology, Hepatology, & Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jing Huang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Diana L Cousminer
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, PA
| | - Amit Patel
- Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Yuwen Xu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Struan F A Grant
- Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, Georgia, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, PA
| | - Robert N Baldassano
- Division of Gastroenterology, Hepatology, & Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lindsey Albenberg
- Division of Gastroenterology, Hepatology, & Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Babette S Zemel
- Division of Gastroenterology, Hepatology, & Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ronen Stein
- Division of Gastroenterology, Hepatology, & Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Fu J, Zhang C. The research progress on radionuclides in osteoporosis diagnosis and drug efficacy monitoring. Front Pharmacol 2025; 16:1594903. [PMID: 40356996 PMCID: PMC12066535 DOI: 10.3389/fphar.2025.1594903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoporosis is a common metabolic bone disease that seriously affects the quality of life and health of patients. Traditional diagnostic methods, such as dual energy X-ray absorptiometry (DXA), have limitations in early detection and dynamic monitoring, making it difficult to meet clinical needs. This paper focuses on the potential of radionuclide imaging techniques, such as Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET), in the early diagnosis of osteoporosis. The paper further elaborates on the importance of radionuclides in evaluating the therapeutic effect of osteoporosis drugs. By summarizing current research findings, this paper aims to emphasize the core role of radionuclides in the management of osteoporosis, and provide theoretical basis and practical guidance for optimizing the diagnosis and treatment strategies of bone metabolism diseases in the future.
Collapse
Affiliation(s)
- Jie Fu
- College of Future Technology, Peking University, Beijing, China
| | - Chi Zhang
- College of Future Technology, Peking University, Beijing, China
- Department of Orthopedics, Peking University International Hospital, Beijing, China
| |
Collapse
|
13
|
Xiang R, Zhao X, Sha L, Tang M, Wu X, Zhang L, Hou J, Deng Q, Qu Y, Zhu J, Qin C, Xiao C, Xiao J, Zhong Y, Yang B, Song X, Zhou J, Han T, Zheng S, Yu T, Liao J, Fan M, Li J, Liu Z, Jiang X. Mapping the role of macro and micronutrients in bone mineral density: a comprehensive Mendelian randomization study. Eur J Nutr 2025; 64:156. [PMID: 40240651 PMCID: PMC12003484 DOI: 10.1007/s00394-025-03665-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 03/22/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Macro and micronutrients may play an important role in osteoporosis development; however, observational studies have yielded inconsistent results. Clarifying these associations is vital to the development of nutritional recommendations aimed at preventing osteoporosis. METHODS Utilizing the largest available genome-wide association study (GWAS) summary statistics to date, we performed a two-sample Mendelian randomization (MR) analysis to investigate the causal effects of energy-adjusted macronutrient intake (fat, protein, carbohydrate, and sugar) and circulating levels of 20 micronutrients (ten each for vitamins and minerals) on heel estimated bone mineral density (eBMD), a promising marker for osteoporosis risk and fracture susceptibility. Sensitivity, sex-specific, and one-sample MR analyses were applied to further validate and annotate the results. RESULTS Among all nutrients, four genetically predicted circulating levels of micronutrients were suggestively associated with eBMD (vitamin A: β IVW = - 0.054, PIVW = 3.70 × 10-2; vitamin B12: β IVW = - 0.020, PIVW = 3.71 × 10-6; vitamin E: β IVW = 0.277, PIVW = 3.22 × 10-2; selenium: β IVW = 0.023, PIVW = 5.37 × 10-3; all Pintercept > 0.05). All these results were also considered robust, as sensitivity analyses yielded directionally consistent results. However, only the causal effects of vitamin B12 and selenium on eBMD remained significant after Bonferroni correction and were not confounded by obesity, smoking status, or alcohol consumption. Sex-specific analysis revealed a male-specific causal association between vitamin E and eBMD ( β IVW = 0.275, PIVW = 9.81 × 10-14). Additionally, using individuallevel data from the UK Biobank cohort, one-sample MR analysis found no causal relationships between diet-derived nutrient intake and eBMD in the overall population, as well as in the females or the males. CONCLUSIONS Our findings suggest that appropriate levels of plasma vitamin B12 and adequate levels of serum selenium are crucial for delaying bone loss and promoting bone health, emphasizing the need for nutritional recommendations to maintain optimal levels of these nutrients to promote eBMD and prevent osteoporosis.
Collapse
Affiliation(s)
- Rong Xiang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xunying Zhao
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Linna Sha
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingshuang Tang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueyao Wu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaojiao Hou
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qin Deng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Qu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiangbo Zhu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenjiarui Qin
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changfeng Xiao
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinyu Xiao
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangdan Zhong
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Yang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Song
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinyu Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Han
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sirui Zheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Yu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaqiang Liao
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mengyu Fan
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayuan Li
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhonghua Liu
- Department of Clinical Nutrition, No.4 West China Teaching Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Xia Jiang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.
| |
Collapse
|
14
|
Zhao X, Li J, Yu J, Shi Y, Tang M. The Role of Sex Steroid Hormones in the Association Between Manganese Exposure and Bone Mineral Density: National Health and Nutrition Examination Survey 2013-2018. TOXICS 2025; 13:296. [PMID: 40278612 PMCID: PMC12031611 DOI: 10.3390/toxics13040296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
This study investigates the association between blood Mn and bone mineral density (BMD), focusing on the mediating role of sex steroids, using data from 8617 participants in the National Health and Nutrition Examination Survey (NHANES) 2013-2018. Weighted multiple linear regression models were used to examine the association of blood Mn and total BMD, and mediation analyses were used to explored the roles of total testosterone (TT), estradiol (E2), and sex hormone-binding globulin (SHBG) in the Mn-BMD relationship, stratified by sex and menopausal status. Blood Mn was negatively associated with BMD in both sexes, with a pronounced effect in postmenopausal women. SHBG mediated 37.16% of the Mn-BMD association in men, whereas no mediating effects were found in women. E2 exhibited a significant indirect effect, suggesting that reduced E2 levels may amplify Mn's effect on BMD. These findings indicate that Mn exposure is associated with decreased BMD, potentially through alterations in sex steroids, highlighting the importance of considering hormone status when evaluating the impact of Mn exposure on BMD.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
- Orthopaedics Research Institute of Zhejiang University, Hangzhou 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Jiayi Li
- Department of Public Health, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jincong Yu
- Department of Orthopeadics, YuYao People’s Hospital, Ningbo 315400, China
| | - Yinhui Shi
- Department of Orthopeadics, CHC International Hospital, Ningbo 315300, China
| | - Mengling Tang
- Department of Public Health, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
15
|
Yang Q, Huang L, Xu H, Feng J, Liu D, Wei S, Jiang H. Gastroesophageal reflux disease and osteoporosis: A bidirectional Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42083. [PMID: 40193675 PMCID: PMC11977714 DOI: 10.1097/md.0000000000042083] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
In observational studies, associations between osteoporosis (OP) and gastroesophageal reflux disease (GERD) have been found. We conducted a 2-way, 2-sample Mendelian randomization (MR) analysis to determine whether these associations have a causal relationship. Data on GERD at the summary-level were sourced from extensive genome-wide association studies encompassing 129,080 cases and 473,524 control subjects. Bone mineral density (BMD) served as the phenotypic indicator for OP. BMD metrics were compiled from a cohort of 537,750 individuals, encompassing total body BMD (TB-BMD) and stratified TB-BMD across age groups, along with BMD measurements at 4 anatomical locations: lumbar spine, femoral neck, heel, and ultra-distal forearm. Multiple MR approaches, such as the inverse-variance weighted (IVW) method, MR-Egger regression, and the MR-PRESSO test, were employed, among which findings obtained by IVW method were designated as the primary outcomes. For quality assurance, sensitivity analyses were conducted using the MR-Egger intercept, Cochran Q, and leave-one-out test. There were no significant causal links between genetic inclination towards GERD and reduced BMD levels. Nonetheless, the genetic evidence suggests a causal link between higher BMD levels and lower incidence of GERD [TB-BMD: OR = 0.941, 95% confidence intervals (CI) = 0.910-0.972, P < .001; TB-BMD-1: OR = 0.919, 95% CI = 0.885-0.954, P < .001; TB-BMD-3: OR = 0.945, 95% CI = 0.915-0.977, P = .001; TB-BMD-4: OR = 0.926, 95% CI = 0.896-0.957, P < .001]. Sensitivity analyses corroborate our findings. The MR analysis indicates no significant causal link between genetic inclination towards GERD and OP or reduced BMD within the European demographic. In addition, the study suggests that lower BMD or OP, as predicted by genetics, may contribute to the development of GERD.
Collapse
Affiliation(s)
- Qinghua Yang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Longao Huang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hongyuan Xu
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junfei Feng
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dun Liu
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shengwang Wei
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker’s Hospital, Liuzhou, China
| | - Hua Jiang
- Department of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
16
|
Wu Z, Yang J, Zhu Y, Li J, Xu K, Li Y, Zhong G, Xu Y, Guo Y, Zhang Y. Causal Associations of Inflammatory Cytokines With Osteosarcopenia: Insights From Mendelian Randomization and Single Cell Analysis. Mediators Inflamm 2025; 2025:6005225. [PMID: 40224485 PMCID: PMC11986192 DOI: 10.1155/mi/6005225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/01/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Osteosarcopenia, the coexistence of osteoporosis and sarcopenia, poses significant challenges in aging populations due to its dual impact on bone and muscle health. Inflammation, mediated by specific cytokines, is thought to play a crucial role in the development of osteosarcopenia, though the underlying mechanisms are not fully understood. Objective: This study aimed to clarify the causal role of circulating cytokines in the pathogenesis of osteosarcopenia by employing mendelian randomization (MR) and single-cell RNA sequencing (scRNA-seq) to identify cell-specific cytokine expression patterns. The ultimate objective was to uncover potential pathological mechanisms and therapeutic targets for treating osteosarcopenia. Methods: A two-sample MR approach was employed, leveraging publicly available genome-wide association study (GWAS) data from multiple cohorts. A total of 91 circulating cytokines were examined using genetic instruments, and their causal effects on traits related to osteoporosis and sarcopenia were evaluated. Various complementary and sensitivity analyses were performed to ensure robust findings. Additionally, scRNA-seq datasets from human muscle and bone marrow were analyzed to validate the single-cell expression profiles of candidate cytokines. Results: MR analysis identified several cytokines with causal effects on osteosarcopenia traits, including LTA, CD40, CXCL6, CXCL10, DNER (delta and notch-like epidermal growth factor-related receptor), and VEGFA (vascular endothelial growth factor A). LTA and CD40 were protective for both bone and muscle, while VEGFA posed a risk. Other cytokines demonstrated opposite effects on bone and muscle. Single cell analysis revealed distinct expression patterns, with LTA highly expressed in lymphocytes, CD40 in immune cells, and VEGFA in various musculoskeletal cell types. Age-related differences in cytokine expression were also noted, with LTA more highly expressed in younger individuals, and VEGFA in older individuals. Conclusion: This study offers preliminary insights into the inflammatory mechanisms potentially driving osteosarcopenia, identifying key cytokines that may be involved in its pathogenesis. By integrating MR and scRNA-seq data, we highlight potential therapeutic targets, though further research is needed to confirm these findings and their implications for musculoskeletal health.
Collapse
Affiliation(s)
- Zugui Wu
- Department of Bone Tumor, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, Guangdong, China
- Department of Orthopaedic, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650000, Yunnan, China
| | - Jiyong Yang
- Department of Orthopaedic, The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong, China
- Department of Orthopaedic, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen Research Institute of Guangzhou University of Traditional Medicine (Futian), Shenzhen 518000, Guangdong, China
| | - Yue Zhu
- Department of Orthopaedic, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650000, Yunnan, China
| | - Jiao Li
- Department of Orthopaedic, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650000, Yunnan, China
| | - Kang Xu
- Department of Orthopaedic, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650000, Yunnan, China
| | - Yuanlong Li
- Department of Bone Tumor, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, Guangdong, China
| | - Guoqing Zhong
- Department of Bone Tumor, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510000, Guangdong, China
| | - Yanfei Xu
- Department of Orthopaedic, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650000, Yunnan, China
| | - Ying Guo
- Department of Orthopaedic, The Third Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650000, Yunnan, China
| | - Yu Zhang
- Department of Bone Tumor, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, Guangdong, China
| |
Collapse
|
17
|
Yang X, Xiao R, Liu B, Xie B, Yang Z. The causal relationship of inflammation-related factors with osteoporosis: A Mendelian Randomization Analysis. Exp Gerontol 2025; 202:112715. [PMID: 39983802 DOI: 10.1016/j.exger.2025.112715] [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: 11/19/2024] [Revised: 02/10/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND We used Mendelian randomization (MR) approach to examine whether genetically determined inflammation-related risk factors play a role in the onset of osteoporosis (OP) in the European population. METHODS Genome-wide association studies (GWASs) summary statistics of estimated bone mineral density (eBMD) obtained from the public database GEnetic Factors for OSteoporosis Consortium (GEFOS) including 142,487 European people. For exposures, we utilized GWAS data of 9 risk factors including diseases chronic kidney disease (CKD) (41,395 cases and 439,303 controls), type 2 diabetes (T2D) (88,427 cases and 566,778 controls), Alzheimer's disease (AD) (71,880 cases, 383,378 controls) and major depression disorder (MDD) (9240 cases and 9519 controls) and lifestyle behaviors are from different consortiums. Inverse variance weighted (IVW) analysis was principal method in this study and random effect model was applied; MR-Egger method and weighted median method were also performed for reliable results. Cochran's Q test and MR-Egger regression were used to detect heterogeneity and pleiotropy and leave-one-out analysis was performed to find out whether there are influential SNPs. RESULTS We found that T2D (IVW: β = 0.05, P = 0.0014), FI (IVW: β = -0.22, P < 0.001), CKD (IVW: β = 0.02, P = 0.009), ALZ (IVW: β = 0.06, P = 0.005), Coffee consumption (IVW: β = 0.11, P = 0.003) were causally associated with OP (P<0.006after Bonferroni correction). CONCLUSIONS Our study revealed that T2D, FI, CKD, ALZ and coffee consumption are causally associated with OP. Future interventions targeting factors above could provide new clinical strategies for the personalized prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Xinyue Yang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Rui Xiao
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, China
| | - Bo Xie
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China.
| | - Zhao Yang
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, China.
| |
Collapse
|
18
|
Du S, Chen J, Li J, Qian W, Wu S, Peng Q, Liu Y, Pan T, Li Y, Hadi SS, Tan J, Yuan Z, Wang J, Tang K, Wang Z, Wen Y, Dong X, Zhou W, Ruiz-Linares A, Shi Y, Jin L, Liu F, Zhang M, Wang S. A multi-ancestry GWAS meta-analysis of facial features and its application in predicting archaic human features. J Genet Genomics 2025; 52:513-524. [PMID: 39002897 DOI: 10.1016/j.jgg.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Facial morphology, a complex trait influenced by genetics, holds great significance in evolutionary research. However, due to limited fossil evidence, the facial characteristics of Neanderthals and Denisovans have remained largely unknown. In this study, we conduct a large-scale multi-ethnic meta-analysis of the genome-wide association study (GWAS), including 9674 East Asians and 10,115 Europeans, quantitatively assessing 78 facial traits using 3D facial images. We identify 71 genomic loci associated with facial features, including 21 novel loci. We develop a facial polygenic score (FPS) that enables the prediction of facial features based on genetic information. Interestingly, the distribution of FPSs among populations from diverse continental groups exhibits relevant correlations with observed facial features. Furthermore, we apply the FPS to predict the facial traits of seven Neanderthals and one Denisovan using ancient DNA and align predictions with the fossil records. Our results suggest that Neanderthals and Denisovans likely share similar facial features, such as a wider but shorter nose and a wider endocanthion distance. The decreased mouth width is characterized specifically in Denisovans. The integration of genomic data and facial trait analysis provides valuable insights into the evolutionary history and adaptive changes in human facial morphology.
Collapse
Affiliation(s)
- Siyuan Du
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jieyi Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Center for Molecular Medicine, Pediatrics Research Institute, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jiarui Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Qian
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sijie Wu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ting Pan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yi Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sibte Syed Hadi
- Department of Forensic Sciences, College of Criminal Justice, Naif Arab University for Security Sciences, Riyadh 11452, Kingdom of Saudi Arabia
| | - Jingze Tan
- Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ziyu Yuan
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu 225326, China
| | - Jiucun Wang
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu 225326, China; Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200120, China; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai 200438, China
| | - Kun Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yanqin Wen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xinran Dong
- Center for Molecular Medicine, Pediatrics Research Institute, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wenhao Zhou
- Center for Molecular Medicine, Pediatrics Research Institute, Children's Hospital of Fudan University, Shanghai 201102, China; Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Andrés Ruiz-Linares
- Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China; Aix-Marseille Université, CNRS, EFS, ADES, Marseille 13005, France; Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Li Jin
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu 225326, China; Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200120, China; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai 200438, China
| | - Fan Liu
- Department of Forensic Sciences, College of Criminal Justice, Naif Arab University for Security Sciences, Riyadh 11452, Kingdom of Saudi Arabia; Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, 3015 CN Rotterdam, the Netherlands
| | - Manfei Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China; Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200120, China.
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| |
Collapse
|
19
|
Mu X, Zhang H, Zhang J. How Accurately Does Bone Mineral Density Predict Bone Strength? A Clinical Observational Study of Osteoporosis Vertebral Compression Fractures in Postmenopausal Women. Orthop Surg 2025; 17:1067-1074. [PMID: 39780751 PMCID: PMC11962276 DOI: 10.1111/os.14354] [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: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES Dual energy x-ray absorptiometry (DXA) provides incomplete information about bone strength. There are few data on the relationship between osteoporosis-related examinations and bone strength. The objective of the present study was to determine which osteoporosis-related examinations best predicted trabecular bone strength, and to enhance a formula for predicting bone strength on the basis of bone density examination. METHODS This observational study included postmenopausal women (aged over 50 years) who underwent unilateral percutaneous kyphoplasty (PKP) surgery in the lumbar spine between September 2021 and June 2023. The pressure within each balloon expansion circle was extracted to reflect the true bone strength. The NHANES 2013-2014 data were used to assess the performance of the formula. The performance of the formula was compared with that of the observed actual fractures. Bland-Altman analysis was used to compare the agreement between the formula and the fracture risk assessment tool (FRAX) score. RESULTS A total of 40 postmenopausal women (mean age ± standard deviation, 70.90 years ± 10.30) were enrolled. The average balloon pressure was 59.23 psi (± 12.40, means ± SDs). The mean BMD of total lumbar spine (average of L1-L4) was 0.89 g/cm2 ± 0.20 (mean ± standard), and the Pearson correlation coefficient between lumbar BMD and bone strength was 0.516. After adjusting for age and BMI, the DXA response rate to bone strength reached 72%. Calibration plots of the observed actual fractures versus those estimated via the bone strength formula were considered good fits. The Bland-Altman analysis revealed a nonsignificant difference between the formula and the FRAX score in predicting fracture risk. CONCLUSIONS After adjustment, the DXA response rate to bone strength reached 72%, indicating a strong correlation. In addition, Bone Strength = DXA × 27 - Age × 0.585-BMI × 0.887 + 98.
Collapse
Affiliation(s)
- Xuemeng Mu
- Department of Orthopedic SurgeryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Hengyan Zhang
- Department of Orthopedic SurgeryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| | - Jia Zhang
- Department of Orthopedic SurgeryPeking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
20
|
Patiño-Salazar JD, Ovejero D, Gabernet M, Martínez-Gil N, Alcaide-Consuegra E, Mellibovsky L, Nogués X, Grinberg D, Balcells S, Rabionet R, Garcia-Giralt N. Identifying rare variants in genes related to bone phenotypes in a cohort of postmenopausal women. Osteoporos Int 2025; 36:637-644. [PMID: 39915337 PMCID: PMC12064449 DOI: 10.1007/s00198-025-07413-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/24/2025] [Indexed: 05/10/2025]
Abstract
Rare genetic variants in genes previously described to be involved in bone monogenic disorders were identified in postmenopausal women split into two groups according to extreme bone mineral density (BMD) values and lumbar spine Z-scores. A pathogenic variant in COL1A2 gene found in a woman with low BMD highlights the overlap between osteogenesis imperfecta and osteoporosis, which may share their genetic etiology. Other variants were not clearly associated with the extreme BMD, suggesting that there is little contribution of rare variants to postmenopausal osteoporosis. PURPOSE We aimed to evaluate whether extreme values of bone mineral density (BMD) in a population-based cohort of postmenopausal women (BARCOS) could be determined by rare genetic variants in genes related to monogenic bone disorders. METHODS A panel of 127 genes related to different skeletal phenotypes was designed. Massive sequencing by targeted capture of these genes was performed in 104 DNA samples from those women of the BARCOS cohort that exhibited the highest (HZ group) and lowest (LZ group) LS Z-scores, ranging from + 0.70 to + 3.80 and from - 2.35 to - 4.26, respectively. 5'UTR, 3'UTR, splice region, missense, nonsense, and short indel variants with MAF < 0.01 were annotated with CADD version 1.6 and considered in the analysis. RESULTS After filtering those variants with CADD > 25 and present only in one of the groups (either LZ or HZ), six variants were detected, most of which (5/6) were in the LZ group in TCIRG1, COL1A2, SEC24D, LRP4, and ANO5 genes, while only one, in the LMNA gene, was in the HZ group. According to the ClinVar database, the COL1A2 variant, causative of a recessive form of osteogenesis imperfecta, is described as pathogenic, while the other variants are considered of uncertain significance (VUS). CONCLUSION The variant identified in COL1A2 in a woman from the LZ group highlights the genetic overlap between monogenic diseases such as osteogenesis imperfecta and complex diseases like osteoporosis. However, the other variants were not clearly associated with the extreme BMD, suggesting that there is little contribution of rare variants to postmenopausal osteoporosis.
Collapse
Affiliation(s)
- J D Patiño-Salazar
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - D Ovejero
- Musculoskeletal Research Group, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - M Gabernet
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - N Martínez-Gil
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - E Alcaide-Consuegra
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - L Mellibovsky
- Musculoskeletal Research Group, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - X Nogués
- Musculoskeletal Research Group, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - D Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - S Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain.
| | - R Rabionet
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - N Garcia-Giralt
- Musculoskeletal Research Group, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| |
Collapse
|
21
|
Chopra A, Song J, Weiner 3rd J, Beule D, Schaefer AS. Genetic analysis of cis-enhancers associated with bone mineral density and periodontitis in the gene SOST. PLoS One 2025; 20:e0319259. [PMID: 40127057 PMCID: PMC11932464 DOI: 10.1371/journal.pone.0319259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/27/2025] [Indexed: 03/26/2025] Open
Abstract
A haplotype block at the sclerostin (SOST) gene correlates with bone mineral density (BMD) and increased periodontitis risk in smokers. Investigating the putative causal variants within this block, our study aimed to elucidate the impact of linked enhancer elements on gene expression and to evaluate their role in transcription factor (TF) binding. Using CRISPR/dCas9 activation (CRISPRa) screening in SaOS-2 cells, we quantified disease-related enhancer activities regulating SOST expression. Additionally, in SaOS-2 cells, we investigated the influence of the candidate TFs CCAAT/enhancer-binding protein beta (CEBPB) on gene expression by antisense (GapmeR) knockdown, followed by RNA sequencing. The periodontitis-linked SNP rs9783823 displayed a significant cis-activating effect (25-fold change in SOST expression), with the C-allele containing a CEBPB binding motif (position weight matrix (PWM) = 0.98, Pcorrected = 7.7 x 10-7). CEBPB knockdown induced genome-wide upregulation but decreased epithelial-mesenchymal transition genes (P = 0.71, AUC = 2.2 x 10-11). This study identifies a robust SOST cis-activating element linked to BMD and periodontitis, carrying CEBPB binding sites, and highlights CEBPB's impact on epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Avneesh Chopra
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jiahui Song
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Arne S. Schaefer
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
22
|
Martínez-Gil N, Herrera-Ubeda C, Gritti N, Roca-Ayats N, Ugartondo N, Garcia-Giralt N, Ovejero D, Nogués X, Garcia-Fernàndez J, Grinberg D, Balcells S. Regulation of WNT16 in bone may involve upstream enhancers within CPED1. Sci Rep 2025; 15:9607. [PMID: 40113825 PMCID: PMC11926113 DOI: 10.1038/s41598-025-93259-z] [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: 09/19/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
WNT16 stands up as an essential gene for bone homeostasis. Here, we present new evidence of the functional role of a particular region within WNT16. Performing 4 C chromatin conformation analysis in three osteoblast-related cells (the human fetal osteoblast hFOB 1.19 cell line, Saos 2 osteosarcoma cell line and mesenchymal Stem Cells -MSC-), we identify physical interactions between the proximal part of WNT16 intron 2, shown here to be an active promoter in Saos 2 osteosarcoma cells, and several putative regulatory regions within CPED1. Analysis of previously published RNA-seq data from hFOB cells disclosed low expression of a region located downstream of this promoter. Our results suggest a novel regulatory mechanism of WNT16 in bone, mediated by physical interaction with various enhancer regions within CPED1.
Collapse
Affiliation(s)
- N Martínez-Gil
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III , Madrid, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - C Herrera-Ubeda
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - N Gritti
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003, Barcelona, Spain
| | - N Roca-Ayats
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III , Madrid, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - N Ugartondo
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III , Madrid, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - N Garcia-Giralt
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Musculoskeletal Research Group, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - D Ovejero
- Musculoskeletal Research Group, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - X Nogués
- Musculoskeletal Research Group, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - J Garcia-Fernàndez
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III , Madrid, Spain.
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain.
| | - Susanna Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
- Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III , Madrid, Spain.
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain.
| |
Collapse
|
23
|
Goovaerts S, Naqvi S, Hoskens H, Herrick N, Yuan M, Shriver MD, Shaffer JR, Walsh S, Weinberg SM, Wysocka J, Claes P. Enhanced insights into the genetic architecture of 3D cranial vault shape using pleiotropy-informed GWAS. Commun Biol 2025; 8:439. [PMID: 40087503 PMCID: PMC11909261 DOI: 10.1038/s42003-025-07875-6] [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: 06/11/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Large-scale GWAS studies have uncovered hundreds of genomic loci linked to facial and brain shape variation, but only tens associated with cranial vault shape, a largely overlooked aspect of the craniofacial complex. Surrounding the neocortex, the cranial vault plays a central role during craniofacial development and understanding its genetics are pivotal for understanding craniofacial conditions. Experimental biology and prior genetic studies have generated a wealth of knowledge that presents opportunities to aid further genetic discovery efforts. Here, we use the conditional FDR method to leverage GWAS data of facial shape, brain shape, and bone mineral density to enhance SNP discovery for cranial vault shape. This approach identified 120 independent genomic loci at 1% FDR, nearly tripling the number discovered through unconditioned analysis and implicating crucial craniofacial transcription factors and signaling pathways. These results significantly advance our genetic understanding of cranial vault shape and craniofacial development more broadly.
Collapse
Affiliation(s)
- Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
| | - Sahin Naqvi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Genetics and Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hanne Hoskens
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
- Department of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research, Institute, University of Calgary, Calgary, AB, Canada
| | - Noah Herrick
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meng Yuan
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Mark D Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - John R Shaffer
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN, USA
| | - Seth M Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anthropology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium.
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| |
Collapse
|
24
|
Zhang G, Zheng J, Zhou Y, Zhou M, Zhang J, Liu Y, Geng Y, Wang W, Xin M, Yang B, Zhang L, Huang L. From teeth to bone: dental caries has causal effects on osteoporosis and osteoporotic fracture. BMC Oral Health 2025; 25:340. [PMID: 40045256 PMCID: PMC11881354 DOI: 10.1186/s12903-025-05735-7] [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: 12/12/2024] [Accepted: 02/28/2025] [Indexed: 03/09/2025] Open
Abstract
OBJECTIVES Evidence from observational studies suggested oral diseases (periodontitis (PD) and dental caries) may increase susceptibility to bone loss. However, inherent confounding of observational studies limits causal interpretation. We aimed to conduct Mendelian randomization (MR) analysis to estimate the causal effect of oral diseases on osteoporosis (OP), bone mineral density (BMD) and fracture risk. METHODS We used summary-level GWAS meta-analysis data from the GLIDE consortium to identify 7 and 17 single-nucleotide polymorphisms (SNPs) for periodontitis and DMFS (the sum of Decayed, Missing, and Filled tooth Surfaces) as the instrumental variables. MR analyses of these instruments were performed on European individuals for the association with BMD of forearm, femoral neck and lumbar spine; and individuals from FinnGen consortium for OP, OP with pathological fracture, postmenopausal OP with pathological fracture, and site-specific fractures. We performed single-variable Mendelian randomization (SVMR) and multivariable Mendelian randomization (MVMR) to simultaneously assess independent causal effects of PD and DMFS on different outcomes. The estimates were primarily derived using inverse variance weighted (IVW) methods. Sensitivity analyses included weighted median, MR-egger, and Leave-one-out test. RESULTS In MVMR, after adjusting for PD, DMFS has a positive causal effect osteoporosis (OR = 1.165, [95% CI 1.020 to 1.331, P = 0.025]) and postmenopausal OP with pathological fracture (OR = 1.422, [95% CI 1.027 to 1.969, P = 0.034]). However, these causal relationships were not observed in the single-variable Mendelian randomization (SVMR) analysis. The causal associations were robust in various sensitivity analyses. CONCLUSIONS In conclusion, dental caries causally increases the risk of OP and postmenopausal OP with pathological fracture, suggesting the existence of teeth-bone axis. Proactive osteoporosis screening in patients with severe dental caries may be warranted for clinical consideration.
Collapse
Affiliation(s)
- Gongzi Zhang
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
- Department of Orthopedics, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Juan Zheng
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Ying Zhou
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Ming Zhou
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Jiali Zhang
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yangxiaoxue Liu
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yuhan Geng
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Wenxin Wang
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Min Xin
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Bo Yang
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Lihai Zhang
- Department of Orthopedics, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.
| | - Liping Huang
- Department of Rehabilitation, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.
| |
Collapse
|
25
|
Michaëlsson K, Zheng R, Baron JA, Fall T, Wolk A, Lind L, Höijer J, Brunius C, Warensjö Lemming E, Titova OE, Svennblad B, Larsson SC, Yuan S, Melhus H, Byberg L, Brooke HL. Cardio-metabolic-related plasma proteins reveal biological links between cardiovascular diseases and fragility fractures: a cohort and Mendelian randomisation investigation. EBioMedicine 2025; 113:105580. [PMID: 39919333 PMCID: PMC11848109 DOI: 10.1016/j.ebiom.2025.105580] [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: 03/05/2024] [Revised: 12/17/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND How cardiovascular diseases (CVD) predispose to a higher risk of fragility fractures is not well understood. Both contribute to significant components of disease burden and health expenditure. Poor bone quality, central obesity, sarcopenia, falls, and low grip strength are independent risk factors for hip and other fragility fractures and also for CVD and early death. METHODS We used proteomics and a cohort design combined with Mendelian randomisation analysis to understand shared mechanisms for developing CVD and fragility fractures, two significant sources of disease burden and health expenditure. We primarily aimed to discover and replicate the association of 274 cardio-metabolic-related proteins with future rates of hip and any fracture in two separate population-based cohorts, with a total of 12,314 women and men. FINDINGS The average age at baseline was 68 years in the discovery cohort of women and 74 years in the mixed-sex replication cohort. During 100,619 person-years of follow-up, 2168 had any fracture, and 538 had a hip fracture. Our analysis resulted in 24 cardiometabolic proteins associated with fracture risk: 20 with hip fracture, 9 with any fracture, and 5 with both. The associations remained even if protein concentrations were measured from specimens taken during preclinical stages of cardio-metabolic diseases, and 19 associations remained after adjustment for bone mineral density. Twenty-two of the proteins were associated with total body fat mass or lean body mass. Mendelian randomisation (MR) analysis supported causality since genetically predicted levels of SOST (Sclerostin), CCDC80 (Coiled-coil domain-containing protein 80), NT-proBNP (N-terminal prohormone brain natriuretic peptide), and BNP (Brain natriuretic peptide) were associated with risk of hip fracture. MR analysis also revealed a possible negative impact on bone mineral density (BMD) by genetically predicted higher levels of SOST, CCDC80, and TIMP4 (Metalloproteinase inhibitor 4). The MR association with BMD was positive for PTX3 (Pentraxin-related protein) and SPP1 (Osteopontin). Genetically predicted higher concentrations of SOST and lower concentrations of SPP1 also conferred a higher risk of falls and lowered grip strength. The genetically determined concentration of nine proteins influenced fat mass, and one influenced lean body mass. INTERPRETATION These data reveal biological links between cardiovascular diseases and fragility fractures. The proteins should be further evaluated as shared targets for developing pharmacological interventions to prevent fractures and cardiovascular disease. FUNDING The study was supported by funding from the Swedish Research Council (https://www.vr.se; grants No. 2015-03257, 2017-00644, 2017-06100, and 2019-01291 to Karl Michaëlsson) and funding from Olle Engkvist Byggmästares stiftelse (SOEB).
Collapse
Affiliation(s)
- Karl Michaëlsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Rui Zheng
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - John A Baron
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Tove Fall
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Alicja Wolk
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Lind
- Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonas Höijer
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Carl Brunius
- Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Eva Warensjö Lemming
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Olga E Titova
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Bodil Svennblad
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susanna C Larsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Melhus
- Clinical Pharmacology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Liisa Byberg
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hannah L Brooke
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
26
|
Lu T, Zhang W, Robinson-Cohen C, Engelman CD, Lu Q, de Boer IH, Sun L, Paterson AD. Characterization of gene-environment interactions for vitamin D through variance quantitative trait loci: a UK Biobank-based genetic epidemiology study. Am J Clin Nutr 2025; 121:731-740. [PMID: 39855341 DOI: 10.1016/j.ajcnut.2025.01.021] [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: 09/05/2024] [Revised: 12/23/2024] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Understanding gene-environment interactions associated with vitamin D status may refine nutrition and public health strategies for vitamin D deficiency. Recent methodological advances have enabled the identification of variance quantitative trait loci (vQTLs) where gene-environment interactions are enriched. OBJECTIVES The study aims to identify vQTLs for serum 25-hydroxy vitamin D (25OHD) concentrations and characterize potential gene-environment interactions of vQTLs. METHODS We conducted vQTL discovery for 25OHD using a newly developed quantile integral linear model in the UK Biobank individuals of European (N = 313,514), African (N = 7800), East Asian (N = 2146), and South Asian (N = 8771) ancestries, respectively. We tested for interactions between the identified vQTL lead variants and 18 environmental, biological, or lifestyle factors, followed by multiple sensitivity analyses. RESULTS We identified 19 independent vQTL lead variants (P < 5 × 10-8) in the European ancestry population. No vQTLs were identified in the non-European ancestry populations, likely because of limited sample sizes. A total of 32 interactions were detected with a false discovery rate <0.05. Although known gene-season of measurement interactions were confirmed, additional interactions were identified involving modifiable risk factors, including time spent outdoors and body mass index. The magnitudes of these interactions were consistent within each locus upon adjusting for the season of measurement and other covariates. We also identified a gene-sex interaction at a vQTL that implicates DHCR7. Integrating transcript- and protein-level evidence, we found that the sex-differentiated genetic associations may act through sex-biased expression of DHCR7 isoforms in skin tissues because of alternative splicing. CONCLUSIONS Through the lens of vQTLs, we identified additional gene-environment interactions affecting vitamin D status in addition to the season of measurement. These findings may provide new insights into the etiology of vitamin D deficiency and encourage personalized prevention and management of associated diseases for at-risk individuals.
Collapse
Affiliation(s)
- Tianyuan Lu
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, United States; Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States; Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada.
| | | | - Cassianne Robinson-Cohen
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Corinne D Engelman
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States; Department of Statistics, University of Wisconsin-Madison, Madison, WI, United States; Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI, United States
| | - Ian H de Boer
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, United States; Kidney Research Institute, University of Washington, Seattle, WA, United States
| | - Lei Sun
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada; Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Andrew D Paterson
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada; Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada; Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
27
|
Loh NY, Vasan SK, Rosoff DB, Roberts E, van Dam AD, Verma M, Phillips D, Wesolowska-Andersen A, Neville MJ, Noordam R, Ray DW, Tobias JH, Gregson CL, Karpe F, Christodoulides C. LRP5 promotes adipose progenitor cell fitness and adipocyte insulin sensitivity. COMMUNICATIONS MEDICINE 2025; 5:51. [PMID: 40000740 PMCID: PMC11862225 DOI: 10.1038/s43856-025-00774-1] [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: 03/04/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND WNT signaling plays a key role in postnatal bone formation. Individuals with gain-of-function mutations in the WNT co-receptor LRP5 exhibit increased lower-body fat mass and potentially enhanced glucose metabolism, alongside high bone mass. However, the mechanisms by which LRP5 regulates fat distribution and its effects on systemic metabolism remain unclear. This study aims to explore the role of LRP5 in adipose tissue biology and its impact on metabolism. METHODS Metabolic assessments and imaging were conducted on individuals with gain- and loss-of-function LRP5 mutations, along with age- and BMI-matched controls. Mendelian randomization analyses were used to investigate the relationship between bone, fat distribution, and systemic metabolism. Functional studies and RNA sequencing were performed on abdominal and gluteal adipose cells with LRP5 knockdown. RESULTS Here we show that LRP5 promotes lower-body fat distribution and enhances systemic and adipocyte insulin sensitivity through cell-autonomous mechanisms, independent of its bone-related functions. LRP5 supports adipose progenitor cell function by activating WNT/β-catenin signaling and preserving valosin-containing protein (VCP)-mediated proteostasis. LRP5 expression in adipose progenitors declines with age, but gain-of-function LRP5 variants protect against age-related fat loss in the lower body. CONCLUSIONS Our findings underscore the critical role of LRP5 in regulating lower-body fat distribution and insulin sensitivity, independent of its effects on bone. Pharmacological activation of LRP5 in adipose tissue may offer a promising strategy to prevent age-related fat redistribution and metabolic disorders.
Collapse
Affiliation(s)
- Nellie Y Loh
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Senthil K Vasan
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel B Rosoff
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Emile Roberts
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Andrea D van Dam
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Manu Verma
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel Phillips
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Agata Wesolowska-Andersen
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Matt J Neville
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - David W Ray
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Celia L Gregson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Fredrik Karpe
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Constantinos Christodoulides
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK.
| |
Collapse
|
28
|
Breusegem SY, Houghton J, Romero-Bueno R, Fragoso-Luna A, Kentistou KA, Ong KK, Janssen AFJ, Bright NA, Riedel CG, Perry JRB, Askjaer P, Larrieu D. A multiparametric anti-aging CRISPR screen uncovers a role for BAF in protein synthesis regulation. Nat Commun 2025; 16:1681. [PMID: 39956852 PMCID: PMC11830792 DOI: 10.1038/s41467-025-56916-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/28/2025] [Indexed: 02/18/2025] Open
Abstract
Progeria syndromes are very rare, incurable premature aging conditions recapitulating most aging features. Here, we report a whole genome, multiparametric CRISPR screen, identifying 43 genes that can rescue multiple cellular phenotypes associated with progeria. We implement the screen in fibroblasts from Néstor-Guillermo Progeria Syndrome male patients, carrying a homozygous A12T mutation in BAF. The hits are enriched for genes involved in protein synthesis, protein and RNA transport and osteoclast formation and are validated in a whole-organism Caenorhabditis elegans model. We further confirm that BAF A12T can disrupt protein synthesis rate and fidelity, which could contribute to premature aging in patients. This work highlights the power of multiparametric genome-wide suppressor screens to identify genes enhancing cellular resilience in premature aging and provide insights into the biology underlying progeria-associated cellular dysfunction.
Collapse
Affiliation(s)
- Sophia Y Breusegem
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Sophia Y. Breusegem: MRC toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Jack Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Jack Houghton: Imperial College London, Exhibition Road, South Kensington, London, UK
| | - Raquel Romero-Bueno
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Adrián Fragoso-Luna
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Katherine A Kentistou
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Anne F J Janssen
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
- Anne F. J. Janssen: Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen, The Netherlands
| | - Nicholas A Bright
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK
| | | | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Peter Askjaer
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas-Universidad Pablo de Olavide-Junta de Andalucía, Seville, Spain
| | - Delphine Larrieu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, UK.
- Delphine Larrieu: Altos Labs, Cambridge Institute of Science, Cambridge, UK.
| |
Collapse
|
29
|
Chen L, Qu Y, Cui H, Zhang W, Wu X, Zhao X, Xiao J, Tang M, Wang Y, Zou Y, Qiu L, Tan Z, Lei B, Ma X, Zhang D, Liu Y, Fan M, Li J, Zhang B, Jiang X. Genomic correlation, shared loci, and causal association between obesity, periodontitis and tooth loss. Sci Rep 2025; 15:5155. [PMID: 39934647 PMCID: PMC11814315 DOI: 10.1038/s41598-025-89782-8] [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: 09/13/2024] [Accepted: 02/07/2025] [Indexed: 02/13/2025] Open
Abstract
Observational studies have reported an association of obesity with periodontitis and tooth loss, yet findings remain inconsistent. We aim to investigate the genetic link underlying obesity-related traits (BMI [body mass index], WHR [waist-to-hip ratio], WHRadjBMI and childhood BMI), periodontitis and tooth loss. Leveraging summary statistics from large-scale genome-wide association studies, we comprehensively investigated the pair-wise genetic correlation using linkage disequilibrium score regression (LDSC) and SUPERGNOVA, identified shared loci using cross-phenotype association analysis (CPASSOC), and estimated causal association using Mendelian randomization. We identified a significant genetic correlation of obesity with tooth loss, but not with periodontitis. Partitioning the genome into LD-independent regions revealed 10 significantly shared local signals across six regions. Genome-wide cross-trait analysis uncovered 14 shared loci, four of which were colocalized: rs2064044 (PP4 = 0.94), rs6000329 (PP4 = 0.86), rs7134628 (PP4 = 0.86), and rs1286769 (PP4 = 0.90). Notably, rs1286769, identified via CPASSOC and validated through colocalization analysis, is located near RARβ, a gene associated with both BMI and denture use. Mendelian randomization revealed a nominally-significant causal association of obesity with periodontitis (P = 0.045) but a robust causal association with tooth loss represented by number of teeth (BMI: beta = [Formula: see text]0.20, 95%CI = [Formula: see text]0.26 to [Formula: see text]0.14, P = 5.27 × 10-11; WHR: beta = [Formula: see text]0.16, 95%CI = [Formula: see text]0.24 to [Formula: see text]0.08, P = 3.71 × 10-5). Results of CAUSE were consistent with main findings. From a genetic perspective, our work highlights an intrinsic link between obesity, periodontitis and tooth loss, which may add new lines of evidence and provide insights for clinical and public oral health applications.
Collapse
Affiliation(s)
- Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yang Qu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Xuan Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Jinyu Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Lingli Qiu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Zhixin Tan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Bowen Lei
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Xiaofeng Ma
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Di Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Mengyu Fan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Ben Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China.
- Departments of Cardiology, Neurology, and Oncology, Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China.
- Department of Occupational and Environmental Health, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China.
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
30
|
Liu R, Fan W, Hu J, Xu K, Huang Z, Liu Y, Sun C. The mediating role of thyroid-related hormones between thyroid dysfunction diseases and osteoporosis: a mediation mendelian randomization study. Sci Rep 2025; 15:4121. [PMID: 39901040 PMCID: PMC11791035 DOI: 10.1038/s41598-025-88412-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
The links between Thyroid dysfunction diseases (TDFDs) and osteoporosis (OP) has received widespread attention, but the causal relationships and mediating factors have not been systematically studied. We used two-sample Mendelian randomization (MR) analysis to elucidate the causal relationship between TDFDs and OP. Moreover, we performed mediation MR analyses to explore the role of thyroid-related hormones and OP risk factors in the association between TDFDs and OP. Two sample MR analyses showed that hyperthyroidism increased OP (OR = 1.080, 95% CI 1.026 to 1.137; P = 0.0032) risk. Hypothyroidism increases OP (OR = 1.183, 95% CI 1.125 to 1.244; P < 0.0001) risk. Furthermore, mediation analysis revealed that TSH mediated 5.314% of the relationship between hypothyroidism and OP. In contrast, FT4 mediated 9.670% of the relationship between hyperthyroidism and OP. In European populations, TDFDs may increase OP risk. TSH mediates in the causal association between hypothyroidism and OP, and similarly, FT4 mediates in the causal link between hyperthyroidism and OP. Our findings underscore the significance of improving integrative care for individuals with TDFDs to mitigate the risk of OP. It is essential to maintain stable levels of thyroid hormones and closely monitor bone health to effectively mitigate and prevent OP.
Collapse
Affiliation(s)
- Ruikang Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Weiming Fan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaming Hu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ke Xu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Zhijian Huang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Liu
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
- Hubei Sizhen Laboratory, Hubei, China.
- Affiliated Hospital of Hubei University of Chinese Medicine, Hubei, China.
| | - Chiyun Sun
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
31
|
Faber BG, Frysz M, Zheng J, Lin H, Flynn KA, Ebsim R, Saunders FR, Beynon R, Gregory JS, Aspden RM, Harvey NC, Lindner C, Cootes T, Evans DM, Davey Smith G, Gao X, Wang S, Kemp JP, Tobias JH. The genetic architecture of hip shape and its role in the development of hip osteoarthritis and fracture. Hum Mol Genet 2025; 34:207-217. [PMID: 39574169 PMCID: PMC11792254 DOI: 10.1093/hmg/ddae169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVES Hip shape is thought to be an important causal risk factor for hip osteoarthritis and fracture. We aimed to identify genetic determinants of hip shape and use these to assess causal relationships with hip osteoarthritis. METHODS Statistical hip shape modelling was used to derive 10 hip shape modes (HSMs) from DXA images in UK Biobank and Shanghai Changfeng cohorts (ntotal = 43 485). Genome-wide association study meta-analyses were conducted for each HSM. Two-sample Mendelian randomisation (MR) was used to estimate causal effects between HSM and hip osteoarthritis using hip fracture as a positive control. RESULTS Analysis of the first 10 HSMs identified 203 independent association signals (P < 5 × 10-9). Hip shape SNPs were also associated (P < 2.5 × 10-4) with hip osteoarthritis (n = 26) and hip fracture (n = 4). Fine mapping implicated SMAD3 and PLEC as candidate genes that may be involved in the development of hip shape and hip osteoarthritis. MR analyses suggested there was no causal effect between any HSM and hip osteoarthritis, however there was evidence that HSM2 (more obtuse neck-shaft angle) and HSM4 (wider femoral neck) have a causal effect on hip fracture (ORIVW method 1.27 [95% CI 1.12-1.44], P = 1.79 × 10-4 and ORIVW 0.74 [0.65-0.84], P = 7.60 × 10-6 respectively). CONCLUSIONS We report the largest hip shape GWAS meta-analysis that identifies hundreds of novel loci, some of which are also associated with hip osteoarthritis and hip fracture. MR analyses suggest hip shape may not cause hip osteoarthritis but is implicated in hip fractures. Consequently, interventions targeting hip shape in older adults to prevent hip osteoarthritis may prove ineffective.
Collapse
Affiliation(s)
- Benjamin G Faber
- Musculoskeletal Research Unit, Learning and Research Building, University of Bristol, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, Oakfield House, University of Bristol, Bristol BS8 2BN, United Kingdom
| | - Monika Frysz
- Musculoskeletal Research Unit, Learning and Research Building, University of Bristol, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, Oakfield House, University of Bristol, Bristol BS8 2BN, United Kingdom
| | - Jaiyi Zheng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Xuhui District, Shanghai 200031, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Kaitlyn A Flynn
- Mater Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane QLD 4102, Australia
| | - Raja Ebsim
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| | - Fiona R Saunders
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Rhona Beynon
- Musculoskeletal Research Unit, Learning and Research Building, University of Bristol, Southmead Hospital, Bristol BS10 5NB, United Kingdom
| | - Jennifer S Gregory
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Richard M Aspden
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Kings College, Aberdeen AB24 3FX, United Kingdom
| | - Nicholas C Harvey
- Medical Research Council Lifecourse Epidemiology Centre, University of Southampton,Tremona Road, Southampton SO16 6YD, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton, Tremona Road, Southampton SO16 6YD, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Claudia Lindner
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| | - Timothy Cootes
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| | - David M Evans
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, Brisbane St Lucia QLD 4067, Australia
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane St Lucia, QLD 4072, Australia
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, Oakfield House, University of Bristol, Bristol BS8 2BN, United Kingdom
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 111 Yixueyuan Road, Xuhui District, Shanghai 200031, China
- Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - John P Kemp
- Medical Research Council Integrative Epidemiology Unit, Oakfield House, University of Bristol, Bristol BS8 2BN, United Kingdom
- Mater Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane QLD 4102, Australia
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane QLD 4102, Australia
| | - Jonathan H Tobias
- Musculoskeletal Research Unit, Learning and Research Building, University of Bristol, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, Oakfield House, University of Bristol, Bristol BS8 2BN, United Kingdom
| |
Collapse
|
32
|
Di D, Zhou H, Cui Z, Zhang J, Liu Q, Yuan T, Zhou T, Luo X, Ling D, Wang Q. Early-life tobacco smoke elevating later-life osteoporosis risk: Mediated by telomere length and interplayed with genetic predisposition. J Adv Res 2025; 68:331-340. [PMID: 38431123 PMCID: PMC11785556 DOI: 10.1016/j.jare.2024.02.021] [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: 12/14/2023] [Revised: 02/11/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024] Open
Abstract
INTRODUCTION The growing prevalence of osteoporosis (OP) in an aging global population presents a significant public health concern. Tobacco smoke negatively affects bone turnover, leading to reduced bone mass and heightened OP and fracture risk. However, the impact of early-life tobacco smoke exposure on later-life OP risk remains unclear. OBJECTIVES This study was to explore the effects of early-life tobacco smoke exposure on incident OP risk in later life. The mediating role of telomere length (TL) and the interaction with genetic predisposition were also studied. METHODS Data on in utero tobacco smoke exposure (IUTSE) status and age of tobacco use initiation from the UK Biobank were used to estimate early-life tobacco smoke exposure. Incident OP cases were identified according to health-related records. Linear, Cox, and Laplace regression models were mainly used for data analysis. RESULTS Individuals with IUTSE showed a higher OP risk [hazard ratio (HR): 1.06, 95 % confidence interval (CI): 1.01, 1.11] and experienced earlier OP onset by 0.30 years [50th percentile difference = -0.30, 95 % CI: -0.51, -0.09] compared to those without. Participants initiating tobacco smoke in childhood, adolescence, and adulthood had 1.41 times (95 % CI: 1.23, 1.61), 1.17 times (95 % CI:1.10, 1.24), and 1.14 times (95 % CI: 1.07, 1.20) the risk of OP, respectively, compared to never smokers. They also experienced earlier OP onset by 2.16, 0.95, and 0.71 years, sequentially. The TL significantly mediated the early-life tobacco exposure and OP association. Significant joint and interactive effects were detected between early-life tobacco smoke exposure and genetic elements. CONCLUSIONS Our findings implicate that early-life tobacco smoke exposure elevates the later-life OP risk, mediated by telomere length and interplayed with genetic predisposition. These findings highlight the importance of early-life intervention against tobacco smoke exposure and ageing status for precise OP prevention, especially in individuals with a high genetic risk.
Collapse
Affiliation(s)
- Dongsheng Di
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Haolong Zhou
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhangbo Cui
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jianli Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qian Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tingting Yuan
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tingting Zhou
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiao Luo
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Danyang Ling
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qi Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
33
|
Alvarado K, Tang WJ, Watson CJ, Ahmed AR, Gómez AE, Donaka R, Amemiya C, Karasik D, Hsu YH, Kwon RY. Loss of cped1 does not affect bone and lean tissue in zebrafish. JBMR Plus 2025; 9:ziae159. [PMID: 39776615 PMCID: PMC11701521 DOI: 10.1093/jbmrpl/ziae159] [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: 08/20/2024] [Revised: 10/28/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Human genetic studies have nominated cadherin-like and PC-esterase domain-containing 1 (CPED1) as a candidate target gene mediating bone mineral density (BMD) and fracture risk heritability. Recent efforts to define the role of CPED1 in bone in mouse and human models have revealed complex alternative splicing and inconsistent results arising from gene targeting, making its function in bone difficult to interpret. To better understand the role of CPED1 in adult bone mass and morphology, we conducted a comprehensive genetic and phenotypic analysis of cped1 in zebrafish, an emerging model for bone and mineral research. We analyzed two different cped1 mutant lines and performed deep phenotyping to characterize more than 200 measures of adult vertebral, craniofacial, and lean tissue morphology. We also examined alternative splicing of zebrafish cped1 and gene expression in various cell/tissue types. Our studies fail to support an essential role of cped1 in adult zebrafish bone. Specifically, homozygous mutants for both cped1 mutant alleles, which are expected to result in loss-of-function and impact all cped1 isoforms, exhibited no significant differences in the measures examined when compared to their respective wildtype controls, suggesting that cped1 does not significantly contribute to these traits. We identified sequence differences in critical residues of the catalytic triad between the zebrafish and mouse orthologs of CPED1, suggesting that differences in key residues, as well as distinct alternative splicing, could underlie different functions of CPED1 orthologs in the two species. Our studies fail to support a requirement of cped1 in zebrafish bone and lean tissue, adding to evidence that variants at 7q31.31 can act independently of CPED1 to influence BMD and fracture risk.
Collapse
Affiliation(s)
- Kurtis Alvarado
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98195, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, United States
| | - W Joyce Tang
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98195, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, United States
| | - Claire J Watson
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98195, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, United States
| | - Ali R Ahmed
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98195, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, United States
| | - Arianna Ericka Gómez
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98195, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, United States
| | - Rajashekar Donaka
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 5290002, Israel
| | - Chris Amemiya
- Department of Molecular and Cell Biology and Quantitative and Systems Biology Program, University of California, Merced, CA 95343, United States
| | - David Karasik
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, 5290002, Israel
- Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA 02131, United States
| | - Yi-Hsiang Hsu
- Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA 02131, United States
| | - Ronald Young Kwon
- Department of Orthopaedic Surgery and Sports Medicine, University of Washington School of Medicine, Seattle, WA 98195, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, United States
| |
Collapse
|
34
|
Zhang Y, Li X, Peng P, Qiu Z, Di C, Chen X, Wang N, Chen F, He Y, Liu Z, Zhao F, Zhu D, Dong S, Hu S, Yang Z, Li Y, Guo Y, Yang T. RUNX2 Phase Separation Mediates Long-Range Regulation Between Osteoporosis-Susceptibility Variant and XCR1 to Promote Osteoblast Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413561. [PMID: 39704037 PMCID: PMC11809430 DOI: 10.1002/advs.202413561] [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: 10/24/2024] [Indexed: 12/21/2024]
Abstract
GWASs have identified many loci associated with osteoporosis, but the underlying genetic regulatory mechanisms and the potential drug target need to be explored. Here, a new regulatory mechanism is found that a GWAS intergenic SNP (rs4683184) functions as an enhancer to influence the binding affinity of transcription factor RUNX2, whose phase separation can mediate the long-range chromatin interaction between enhancer and target gene XCR1 (a member of the GPCR family), leading to changes of XCR1 expression and osteoblast differentiation. Bone-targeting AAV of Xcr1 can improve bone formation in osteoporosis mice, suggesting that XCR1 can be a new susceptibility gene for osteoporosis. This study is the first to link non-coding SNP with phase separation, providing a new insight into long-range chromatin regulation mechanisms with susceptibility to complex diseases, and finding a potential target for the development of osteoporosis drugs and corresponding translational research.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Xin‐Hao Li
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Pai Peng
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Zi‐Han Qiu
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Chen‐Xi Di
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Xiao‐Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Nai‐Ning Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Fei Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Yin‐Wei He
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Zhong‐Bo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Fan Zhao
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Dong‐Li Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Shan‐Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Shou‐Ye Hu
- Department of Joint SurgeryHonghui HospitalXi'an Jiaotong UniversityXi'anShaanxi710054China
| | - Zhi Yang
- Department of Joint SurgeryHonghui HospitalXi'an Jiaotong UniversityXi'anShaanxi710054China
| | - Yi‐Ping Li
- Division in Cellular and Molecular MedicineDepartment of Pathology and Laboratory MedicineTulane University School of MedicineTulane UniversityNew OrleansLA70112USA
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Tie‐Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| |
Collapse
|
35
|
Zheng Q, Lin R, Wang D, Chen R, Xu W. The association of lipids and novel non-statin lipid-lowering drug target with osteoporosis: evidence from genetic correlations and Mendelian randomization. BMC Musculoskelet Disord 2025; 26:107. [PMID: 39893413 PMCID: PMC11787747 DOI: 10.1186/s12891-024-08160-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/05/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND It remains controversial whether lipids affect osteoporosis (OP) or bone mineral density (BMD), and causality has not been established. This study aimed to investigate the genetic associations between lipids, novel non-statin lipid-lowering drug target genes, and OP and BMD. METHODS Mendelian randomization (MR) method was used to explore the genetic associations between 179 lipid species and OP, BMD. Drug-target MR analysis was used to explore the causal associations between angiopoietin-like protein 3 (ANGPTL3) and apolipoprotein C3 (APOC3) inhibitors on BMD. RESULTS The IVW results with Bonferroni correction indicated that triglyceride (TG) (51:3) (OR = 1.0029; 95% CI: 1.0014-1.0045; P = 0.0002) and TG (56:6) (OR = 1.0021; 95% CI: 1.0008-1.0033; P = 0.0011) were associated with an increased risk of OP; TG (51:2) (OR = 0.9543; 95% CI: 0.9148-0.9954; P = 0.0298) was associated with decreased BMD; and ANGPTL3 inhibitor (OR = 1.1342; 95% CI: 1.0393-1.2290; P = 0.0093) and APOC3 inhibitor (OR = 1.0506; 95% CI: 1.0155-1.0857; P = 0.0058) was associated with increased BMD. CONCLUSIONS MR analysis indicated causal associations between genetically predicted TGs and OP and BMD. Drug-target MR analysis showed that ANGPTL3 and APOC3 have the potential to serve as novel non-statin lipid-lowering drug targets to treat or prevent OP.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Orthopedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People's Hospital, Beijing, 100044, China
| | - Rongsheng Chen
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Department of Orthopedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China.
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Department of Orthopedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
36
|
Ru Y, Ma M, Zhou X, Kriti D, Cohen N, D'Souza S, Schaniel C, Motch Perrine SM, Kuo S, Pichurin O, Pinto D, Housman G, Holmes G, Schadt E, van Bakel H, Zhang B, Jabs EW, Wu M. Integrated transcriptomic analysis of human induced pluripotent stem cell-derived osteogenic differentiation reveals a regulatory role of KLF16. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.11.579844. [PMID: 38405902 PMCID: PMC10888757 DOI: 10.1101/2024.02.11.579844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Osteogenic differentiation is essential for bone development, metabolism, and repair; however, the underlying regulatory relationships among genes remain poorly understood. To elucidate the transcriptomic changes and identify novel regulatory genes involved in osteogenic differentiation, we differentiated mesenchymal stem cells (MSCs) derived from 20 human iPSC lines into preosteoblasts (preOBs) and osteoblasts (OBs). We then performed transcriptome profiling of MSCs, preOBs and OBs. The iPSC-derived MSCs and OBs showed similar transcriptome profiles to those of primary human MSCs and OBs, respectively. Differential gene expression analysis revealed global changes in the transcriptomes from MSCs to preOBs, and then to OBs, including the differential expression of 840 genes encoding transcription factors (TFs). TF regulatory network analysis uncovered a network comprising 451 TFs, organized into five interactive modules. Multiscale embedded gene co-expression network analysis (MEGENA) identified gene co-expression modules and key network regulators (KNRs). From these analyses, KLF16 emerged as an important TF in osteogenic differentiation. We demonstrate that overexpression of Klf16 in vitro inhibited osteogenic differentiation and mineralization, while Klf16 +/- mice exhibited increased bone mineral density, trabecular number, and cortical bone area. Our study underscores the complexity of osteogenic differentiation and identifies novel regulatory genes such as KLF16, which plays an inhibitory role in osteogenic differentiation both in vitro and in vivo.
Collapse
Affiliation(s)
- Ying Ru
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Meng Ma
- Mount Sinai Genomics, Sema4, Stamford, CT, 06902, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Divya Kriti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Present address: Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC V6T 2G3, Canada
| | - Ninette Cohen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Present address: Division of Cytogenetics and Molecular Pathology, Zucker School of Medicine at Hofstra/Northwell, Northwell Health Laboratories, Lake Success, NY, 11030, USA
| | - Sunita D'Souza
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Present address: St Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Christoph Schaniel
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Susan M Motch Perrine
- Department of Anthropology, Pennsylvania State University, University Park, PA, 16802, USA
| | - Sharon Kuo
- Department of Biomedical Sciences, University of Minnesota, Duluth, MN, 55812, USA
- Technological Primates Research Group, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany
| | - Oksana Pichurin
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Dalila Pinto
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Genevieve Housman
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany
| | - Greg Holmes
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Meng Wu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
37
|
Debaenst S, Jarayseh T, De Saffel H, Bek JW, Boone M, Josipovic I, Kibleur P, Kwon RY, Coucke PJ, Willaert A. Crispant analysis in zebrafish as a tool for rapid functional screening of disease-causing genes for bone fragility. eLife 2025; 13:RP100060. [PMID: 39817421 PMCID: PMC11737869 DOI: 10.7554/elife.100060] [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] [Indexed: 01/18/2025] Open
Abstract
Heritable fragile bone disorders (FBDs), ranging from multifactorial to rare monogenic conditions, are characterized by an elevated fracture risk. Validating causative genes and understanding their mechanisms remain challenging. We assessed a semi-high throughput zebrafish screening platform for rapid in vivo functional testing of candidate FBD genes. Six genes linked to severe recessive osteogenesis imperfecta (OI) and four associated with bone mineral density (BMD) from genome-wide association studies were analyzed. Using CRISPR/Cas9-based crispant screening in F0 mosaic founder zebrafish, Next-generation sequencing confirmed high indel efficiency (mean 88%), mimicking stable knock-out models. Skeletal phenotyping at 7, 14, and 90 days post-fertilization (dpf) using microscopy, Alizarin Red S staining, and microCT was performed. Larval crispants showed variable osteoblast and mineralization phenotypes, while adult crispants displayed consistent skeletal defects, including malformed neural and haemal arches, vertebral fractures and fusions, and altered bone volume and density. In addition, aldh7a1 and mbtps2 crispants experienced increased mortality due to severe skeletal deformities. RT-qPCR revealed differential expression of osteogenic markers bglap and col1a1a, highlighting their biomarker potential. Our results establish zebrafish crispant screening as a robust tool for FBD gene validation, combining skeletal and molecular analyses across developmental stages to uncover novel insights into gene functions in bone biology.
Collapse
Affiliation(s)
- Sophie Debaenst
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent UniversityGhentBelgium
| | - Tamara Jarayseh
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent UniversityGhentBelgium
| | - Hanna De Saffel
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent UniversityGhentBelgium
| | - Jan Willem Bek
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent UniversityGhentBelgium
| | - Matthieu Boone
- Center for X-ray Tomography, Department of Physics and Astronomy, Ghent UniversityGhentBelgium
| | - Ivan Josipovic
- Center for X-ray Tomography, Department of Physics and Astronomy, Ghent UniversityGhentBelgium
| | - Pierre Kibleur
- Center for X-ray Tomography, Department of Physics and Astronomy, Ghent UniversityGhentBelgium
| | - Ronald Y Kwon
- Department of Orthopaedics and Sports Medicine, University of WashingtonSeattleUnited States
- Institute for Stem Cell and Regenerative Medicine, University of WashingtonSeattleUnited States
| | - Paul J Coucke
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent UniversityGhentBelgium
| | - Andy Willaert
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent UniversityGhentBelgium
| |
Collapse
|
38
|
Dong SS, Duan YY, Zhu RJ, Jia YY, Chen JX, Huang XT, Tang SH, Yu K, Shi W, Chen XF, Jiang F, Hao RH, Liu Y, Liu Z, Guo Y, Yang TL. Systematic functional characterization of non-coding regulatory SNPs associated with central obesity. Am J Hum Genet 2025; 112:116-134. [PMID: 39753113 PMCID: PMC11739881 DOI: 10.1016/j.ajhg.2024.11.005] [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: 08/05/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 01/20/2025] Open
Abstract
Central obesity is associated with higher risk of developing a wide range of diseases independent of overall obesity. Genome-wide association studies (GWASs) have identified more than 300 susceptibility loci associated with central obesity. However, the functional understanding of these loci is limited by the fact that most loci are in non-coding regions. To address this issue, our study first prioritized 2,034 single-nucleotide polymorphisms (SNPs) based on fine-mapping and epigenomic annotation analysis. Subsequently, we employed self-transcribing active regulatory region sequencing (STARR-seq) to systematically evaluate the enhancer activity of these prioritized SNPs. The resulting data analysis identified 141 SNPs with allelic enhancer activity. Further analysis of allelic transcription factor (TF) binding prioritized 20 key TFs mediating the central-obesity-relevant genetic regulatory network. Finally, as an example, we illustrate the molecular mechanisms of how rs8079062 acts as an allele-specific enhancer to regulate the expression of its targeted RNF157. We also evaluated the role of RNF157 in the adipogenic differentiation process. In conclusion, our results provide an important resource for understanding the genetic regulatory mechanisms underlying central obesity.
Collapse
Affiliation(s)
- Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yuan-Yuan Duan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Ren-Jie Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ying-Ying Jia
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Jia-Xin Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Xiao-Ting Huang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Shi-Hao Tang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ke Yu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Wei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Feng Jiang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Ruo-Han Hao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Key Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutions, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| |
Collapse
|
39
|
Yang F, Wen J. Association between bone mineral density and scoliosis: a two-sample mendelian randomization study in european populations. Hereditas 2024; 161:57. [PMID: 39736789 DOI: 10.1186/s41065-024-00352-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/19/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Previous studies have shown that bone mineral density (BMD) has a certain impact on scoliosis. However, up to now, there is no clear evidence that there is a causal association between the two. The aim of this study is to investigate whether there is a causal association between BMD at different body positions and scoliosis by two-sample Mendelian randomization (MR). METHODS Genetic variants (SNPS) strongly associated with BMD (total body BMD (TB-BMD), lumbar spine BMD (LS-BMD), femoral neck BMD (FN-BMD), heel BMD (HE-BMD), and forearm BMD (FA-BMD)) were extracted from GEFOS and genome-wide association analysis (GWAS) databases SNPs) were used as instrumental variables (IVs). Scoliosis was also selected from the Finnish database as the outcome. Inverse variance weighting (IVW) method was used as the main analysis method, and multiple sensitivity analysis was performed by combining weighted median, MR-Egger, MR Multi-effect residuals and outliers. RESULTS IVW results showed that TB-BMD (OR = 0.83, 95%CI: 0.66-1.55 P = 0.13), LS-BMD (OR = 0.72, 95%CI: 0.52-0.99, P = 0.04), FN-BMD (OR = 0.74, 95%CI: 0.50-1.09, P = 0.13), FA-BMD (OR = 0.95,95%CI: 0.70-1.28, P = 0.75), HE-BMD (OR = 0.91, 95%CI: 0.77-1.08, P = 0.29). Sensitivity analyses showed no evidence of pleiotropy or heterogeneity (p > 0.05) (MR-PRESSO and Cochrane). The results were further validated by leave-one-out test and MR-Egger intercept, which confirmed the robustness of the study results. CONCLUSION In conclusion, the present study demonstrates that the causal role of genetic prediction of scoliosis increases with decreasing lumbar BMD. There was no evidence that BMD at the remaining sites has a significant causal effect on scoliosis. Our results suggest that the lumbar spine BMD should be routinely measured in the population at high risk of scoliosis. If osteoporosis occurs, appropriate treatment should be given to reduce the incidence of scoliosis. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Fangjun Yang
- Department of orthopedic, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Jiantao Wen
- Department of Pediatric Spine Surgery, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China.
| |
Collapse
|
40
|
Zhou W, van de Laarschot DM, van Rooij JGJ, Koedam M, Nguyen HH, Uitterlinden AG, Ebeling PR, Thakker RV, Geusens P, van der Eerden BCJ, Verkerk AJMH, Zillikens MC. Family-based whole-exome sequencing implicates a variant in lysyl oxidase like 4 in atypical femur fractures. J Bone Miner Res 2024; 40:69-78. [PMID: 39485938 DOI: 10.1093/jbmr/zjae175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/19/2024] [Accepted: 10/31/2024] [Indexed: 11/03/2024]
Abstract
Atypical femur fractures (AFFs) are rare adverse events associated with bisphosphonate use, having unclear pathophysiology. AFFs also cluster in families and have occurred in patients with monogenetic bone diseases sometimes without bisphosphonate use, suggesting an underlying genetic susceptibility. Our aim was to identify a genetic cause for AFF in a Caucasian family with 7 members affected by osteoporosis, including 3 siblings with bisphosphonate-associated AFFs. Using whole-exome sequencing, we identified a rare pathogenic variant c.G1063A (p.Gly355Ser) in lysyl oxidase like 4 (LOXL4) among 64 heterozygous rare, protein-altering variants shared by the 3 siblings with AFFs. The same variant was also found in a fourth sibling with a low-trauma femur fracture above the knee, not fulfilling all the ASBMR criteria of AFF and in 1 of 73 unrelated European AFF patients. LOXL4 is involved in collagen cross-linking and may be relevant for microcrack formation and bone repair mechanisms. Preliminary functional analysis showed that skin fibroblast-derived osteoblasts from the unrelated patient with the LOXL4 variant expressed less collagen type I and elastin, while osteogenic differentiation and mineralization were enhanced compared with 2 controls. In conclusion, this LOXL4 variant may underlie AFF susceptibility possibly due to abnormal collagen metabolism, leading to increased formation of microdamage or compromised healing of microcracks in the femur.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| | - Denise M van de Laarschot
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| | - Jeroen G J van Rooij
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| | - Marijke Koedam
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| | - Hanh H Nguyen
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford, OX3 9DU, United Kingdom
| | - Piet Geusens
- Biomedical Research Institute, University Hasselt, Diepenbeek, 3500, Belgium
- Department of Internal Medicine, Maastricht University, Maastricht, 6211 LK, the Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| | - Annemieke J M H Verkerk
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, 3015 GD, the Netherlands
| |
Collapse
|
41
|
Yang MY, Zhong JD, Li X, Tian G, Bai WY, Fang YH, Qiu MC, Yuan CD, Yu CF, Li N, Yang JJ, Liu YH, Yu SH, Zhao WW, Liu JQ, Sun Y, Cong PK, Khederzadeh S, Zhao PP, Qian Y, Guan PL, Gu JX, Gai SR, Yi XJ, Tao JG, Chen X, Miao MM, Lei LX, Xu L, Xie SY, Li JC, Guo JF, Karasik D, Yang L, Tang BS, Huang F, Zheng HF. SEAD reference panel with 22,134 haplotypes boosts rare variant imputation and genome-wide association analysis in Asian populations. Nat Commun 2024; 15:10839. [PMID: 39738056 PMCID: PMC11686012 DOI: 10.1038/s41467-024-55147-4] [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: 12/06/2023] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Limited whole genome sequencing (WGS) studies in Asian populations result in a lack of representative reference panels, thus hindering the discovery of ancestry-specific variants. Here, we present the South and East Asian reference Database (SEAD) panel ( https://imputationserver.westlake.edu.cn/ ), which integrates WGS data for 11,067 individuals from various sources across 17 Asian countries. The SEAD panel, comprising 22,134 haplotypes and 88,294,957 variants, demonstrates improved imputation accuracy for South Asian populations compared to 1000 Genomes Project, TOPMed, and ChinaMAP panels, with a higher proportion of well-imputed rare variants. For East Asian populations, SEAD shows concordance comparable to ChinaMAP, but outperforming TOPMed. Additionally, we apply the SEAD panel to conduct a genome-wide association study for total hip (Hip) and femoral neck (FN) bone mineral density (BMD) traits in 5369 genotyped Chinese samples. The single-variant test suggests that rare variants near SNTG1 are associated with Hip BMD (rs60103302, MAF = 0.0092, P = 1.67 × 10-7), and variant-set analysis further supports the association (Pslide_window = 9.08 × 10-9, Pgene_centric = 5.27 × 10-8). This association was not reported previously and can only be detected by using Asian reference panels. Preliminary in vitro experiments for one of the rare variants identified provide evidence that it upregulates SNTG1 expression, which could in turn inhibit the proliferation and differentiation of preosteoblasts.
Collapse
Affiliation(s)
- Meng-Yuan Yang
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jia-Dong Zhong
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xin Li
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Geng Tian
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Wei-Yang Bai
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yi-Hu Fang
- WBBC Jiangxi Center, Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Mo-Chang Qiu
- WBBC Jiangxi Center, Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Cheng-Da Yuan
- Department of Dermatology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Chun-Fu Yu
- Department of Orthopedic Surgery, Shangrao Municipal Hospital, Shangrao, Jiangxi, China
| | - Nan Li
- The High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Ji-Jian Yang
- The High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Yu-Heng Liu
- The High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Shi-Hui Yu
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd, Guangzhou, Guangdong, China
| | - Wei-Wei Zhao
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd, Guangzhou, Guangdong, China
| | - Jun-Quan Liu
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd, Guangzhou, Guangdong, China
| | - Yi Sun
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd, Guangzhou, Guangdong, China
| | - Pei-Kuan Cong
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Saber Khederzadeh
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Pian-Pian Zhao
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yu Qian
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Peng-Lin Guan
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jia-Xuan Gu
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Si-Rui Gai
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xiang-Jiao Yi
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jian-Guo Tao
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xiang Chen
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Mao-Mao Miao
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Lan-Xin Lei
- Medical Biosciences, Imperial College London, London, United Kingdom
| | - Lin Xu
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Shu-Yang Xie
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Jin-Chen Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center for Medical Genetics & Hunan Key Laboratory, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Ji-Feng Guo
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center for Medical Genetics & Hunan Key Laboratory, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bei-Sha Tang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center for Medical Genetics & Hunan Key Laboratory, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Fei Huang
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Hou-Feng Zheng
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
42
|
Conery M, Pippin JA, Wagley Y, Trang K, Pahl MC, Villani DA, Favazzo LJ, Ackert-Bicknell CL, Zuscik MJ, Katsevich E, Wells AD, Zemel BS, Voight BF, Hankenson KD, Chesi A, Grant SF. GWAS-Informed data integration and non-coding CRISPRi screen illuminate genetic etiology of bone mineral density. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585778. [PMID: 38562830 PMCID: PMC10983984 DOI: 10.1101/2024.03.19.585778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Over 1,100 independent signals have been identified with genome-wide association studies (GWAS) for bone mineral density (BMD), a key risk factor for mortality-increasing fragility fractures; however, the effector gene(s) for most remain unknown. Informed by a variant-to-gene mapping strategy implicating 89 non-coding elements predicted to regulate osteoblast gene expression at BMD GWAS loci, we executed a single-cell CRISPRi screen in human fetal osteoblasts (hFOBs). The BMD relevance of hFOBs was supported by heritability enrichment from stratified LD-score regression involving 98 cell types grouped into 15 tissues. 23 genes showed perturbation in the screen, with four (ARID5B, CC2D1B, EIF4G2, and NCOA3) exhibiting consistent effects upon siRNA knockdown on three measures of osteoblast maturation and mineralization. Lastly, additional heritability enrichments, genetic correlations, and multi-trait fine-mapping revealed unexpectedly that many BMD GWAS signals are pleiotropic and likely mediate their effects via non-bone tissues. Extending our CRISPRi screening approach to these tissues could play a key role in fully elucidating the etiology of BMD.
Collapse
Affiliation(s)
- Mitchell Conery
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Khanh Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David A. Villani
- Colorado Program for Musculoskeletal Research, University of Colorado Anschutz Medical Campus, Aurora, CO
- Cell Biology, Stems Cells and Development Ph.D. Program, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Lacey J. Favazzo
- Colorado Program for Musculoskeletal Research, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- University of Colorado Interdisciplinary Joint Biology Program, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Cheryl L. Ackert-Bicknell
- Colorado Program for Musculoskeletal Research, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- University of Colorado Interdisciplinary Joint Biology Program, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Michael J. Zuscik
- Colorado Program for Musculoskeletal Research, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- University of Colorado Interdisciplinary Joint Biology Program, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Eugene Katsevich
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Babette S. Zemel
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Benjamin F. Voight
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
43
|
Khan NM, Wilderman A, Kaiser JM, Kamalakar A, Goudy SL, Cotney J, Drissi H. Enhanced osteogenic potential of iPSC-derived mesenchymal progenitor cells following genome editing of GWAS variants in the RUNX1 gene. Bone Res 2024; 12:70. [PMID: 39643619 PMCID: PMC11624199 DOI: 10.1038/s41413-024-00369-x] [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: 03/19/2024] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 12/09/2024] Open
Abstract
Recent genome-wide association studies (GWAS) identified 518 significant loci associated with bone mineral density (BMD), including variants at the RUNX1 locus (rs13046645, rs2834676, and rs2834694). However, their regulatory impact on RUNX1 expression and bone formation remained unclear. This study utilized human induced pluripotent stem cells (iPSCs) differentiated into osteoblasts to investigate these variants' regulatory roles. CRISPR/Cas9 was employed to generate mutant (Δ) iPSC lines lacking these loci at the RUNX1 locus. Deletion lines (Δ1 and Δ2) were created in iPSCs to assess the effects of removing regions containing these loci. Deletion lines exhibited enhanced osteogenic potential, with increased expression of osteogenic marker genes and Alizarin Red staining. Circularized chromosome conformation capture (4C-Seq) was utilized to analyze interactions between BMD-associated loci and the RUNX1 promoter during osteogenesis. Analysis revealed altered chromatin interactions with multiple gene promoters including RUNX1 isoform, as well as SETD4, a histone methyltransferase, indicating their regulatory influence. Interestingly, both deletion lines notably stimulated the expression of the long isoform of RUNX1, with more modest effects on the shorter isoform. Consistent upregulation of SETD4 and other predicted targets within the Δ2 deletion suggested its removal removed a regulatory hub constraining expression of multiple genes at this locus. In vivo experiments using a bone defect model in mice demonstrated increased bone regeneration with homozygous deletion of the Δ2 region. These findings indicate that BMD-associated variants within the RUNX1 locus regulate multiple effector genes involved in osteoblast commitment, providing valuable insights into genetic regulation of bone density and potential therapeutic targets.
Collapse
Affiliation(s)
- Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Andrea Wilderman
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Jarred M Kaiser
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Archana Kamalakar
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven L Goudy
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA.
- Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
44
|
Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, Song G. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024; 9:340. [PMID: 39627201 PMCID: PMC11615378 DOI: 10.1038/s41392-024-02030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
The dynamic regulation of chromatin accessibility is one of the prominent characteristics of eukaryotic genome. The inaccessible regions are mainly located in heterochromatin, which is multilevel compressed and access restricted. The remaining accessible loci are generally located in the euchromatin, which have less nucleosome occupancy and higher regulatory activity. The opening of chromatin is the most important prerequisite for DNA transcription, replication, and damage repair, which is regulated by genetic, epigenetic, environmental, and other factors, playing a vital role in multiple biological progresses. Currently, based on the susceptibility difference of occupied or free DNA to enzymatic cleavage, solubility, methylation, and transposition, there are many methods to detect chromatin accessibility both in bulk and single-cell level. Through combining with high-throughput sequencing, the genome-wide chromatin accessibility landscape of many tissues and cells types also have been constructed. The chromatin accessibility feature is distinct in different tissues and biological states. Research on the regulation network of chromatin accessibility is crucial for uncovering the secret of various biological processes. In this review, we comprehensively introduced the major functions and mechanisms of chromatin accessibility variation in different physiological and pathological processes, meanwhile, the targeted therapies based on chromatin dynamics regulation are also summarized.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Li
- Hepatobiliary Pancreatic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lingli Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China.
| |
Collapse
|
45
|
Li Z, Hao L, Chen S, Fu W, Zhang H, Yin Z, Wang Y, Wang J. Forkhead box C1 promotes the pathology of osteoarthritis in subchondral bone osteoblasts via the Piezo1/YAP axis. Cell Signal 2024; 124:111463. [PMID: 39396563 DOI: 10.1016/j.cellsig.2024.111463] [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: 07/22/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Subchondral bone sclerosis is a key characteristic of osteoarthritis (OA). Prior research has shown that Forkhead box C1 (FoxC1) plays a role in the synovial inflammation of OA, but its specific role in the subchondral bone of OA has not been explored. Our research revealed elevated expression levels of FoxC1 and Piezo1 in OA subchondral bone tissues. Further experiments on OA subchondral bone osteoblasts with FoxC1 or Piezo1 overexpression showed increased cell proliferation activity, expression of Yes-associated Protein 1 (YAP) and osteogenic markers, and secretion of proinflammatory factors. Mechanistically, the overexpression of FoxC1 through Piezo1 activation, in combination with downstream YAP signaling, led to increased levels of alkaline phosphatase (ALP), collagen type 1 (COL1) A1, RUNX2, Osteocalcin, matrix metalloproteinase (MMP) 3, and MMP9 expression. Notably, inhibition of Piezo1 reversed the regulatory function of FoxC1. The binding of FoxC1 to the targeted area (ATATTTATTTA, residues +612 to +622) and the activation of Piezo1 transcription were verified by the dual luciferase assays. Additionally, Reduced subchondral osteosclerosis and microangiogenesis were observed in knee joints from FoxC1-conditional knockout (CKO) and Piezo1-CKO mice, indicating reduced lesions. Collectively, our study reveals the significant involvement of FoxC1 in the pathologic process of OA subchondral bone via the Piezo1/YAP signaling pathway, potentially establishing a novel therapeutic target.
Collapse
Affiliation(s)
- Zhengyuan Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Lin Hao
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Shenghong Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Wenhan Fu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Hui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China
| | - Zongsheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China.
| | - Yin Wang
- Department of Wound Repair & Plastic and Aesthetic Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui, China; Anhui Public Health Clinical Center, Anhui, China.
| | - Jun Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China.
| |
Collapse
|
46
|
Rakowski A, Monti R, Lippert C. TransferGWAS of T1-weighted brain MRI data from UK Biobank. PLoS Genet 2024; 20:e1011332. [PMID: 39671448 DOI: 10.1371/journal.pgen.1011332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/31/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024] Open
Abstract
Genome-wide association studies (GWAS) traditionally analyze single traits, e.g., disease diagnoses or biomarkers. Nowadays, large-scale cohorts such as UK Biobank (UKB) collect imaging data with sample sizes large enough to perform genetic association testing. Typical approaches to GWAS on high-dimensional modalities extract predefined features from the data, e.g., volumes of regions of interest. This limits the scope of such studies to predefined traits and can ignore novel patterns present in the data. TransferGWAS employs deep neural networks (DNNs) to extract low-dimensional representations of imaging data for GWAS, eliminating the need for predefined biomarkers. Here, we apply transferGWAS on brain MRI data from UKB. We encoded 36, 311 T1-weighted brain magnetic resonance imaging (MRI) scans using DNN models trained on MRI scans from the Alzheimer's Disease Neuroimaging Initiative, and on natural images from the ImageNet dataset, and performed a multivariate GWAS on the resulting features. We identified 289 independent loci, associated among others with bone density, brain, or cardiovascular traits, and 11 regions having no previously reported associations. We fitted polygenic scores (PGS) of the deep features, which improved predictions of bone mineral density and several other traits in a multi-PGS setting, and computed genetic correlations with selected phenotypes, which pointed to novel links between diffusion MRI traits and type 2 diabetes. Overall, our findings provided evidence that features learned with DNN models can uncover additional heritable variability in the human brain beyond the predefined measures, and link them to a range of non-brain phenotypes.
Collapse
Affiliation(s)
- Alexander Rakowski
- Digital Health Machine Learning, Hasso Plattner Institute for Digital Engineering, University of Potsdam, Germany
| | - Remo Monti
- Digital Health Machine Learning, Hasso Plattner Institute for Digital Engineering, University of Potsdam, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Christoph Lippert
- Digital Health Machine Learning, Hasso Plattner Institute for Digital Engineering, University of Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
47
|
Reppe S, Reseland JE, Prijatelj V, Prediger M, Nogueira LP, Utheim TP, Rivadeneira F, Gautvik KM, Datta HK. The DLEU2-miR-15a-16-1 Cluster Is a Determinant of Bone Microarchitecture and Strength in Postmenopausal Women and Mice. Int J Mol Sci 2024; 25:12724. [PMID: 39684435 DOI: 10.3390/ijms252312724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
This study explores how select microRNAs (miRNAs) influence bone structure in humans and in transgenic mice. In trabecular bone biopsies from 84 postmenopausal women (healthy, osteopenic, and osteoporotic), we demonstrate that DLEU2 (deleted in lymphocytic leukemia 2)-encoded miR-15a-5p is strongly positively associated with bone mineral density (BMD) at different skeletal sites. In bone transcriptome analyses, miR-15a-5p levels correlated positively with the osteocyte characteristic transcripts SOST (encoding sclerostin) and MEPE (Matrix Extracellular Phosphoglycoprotein), while the related miR-15b-5p showed a negative association with BMD and osteoblast markers. The data imply that these miRNAs have opposite roles in bone remodeling with distinct actions on bone cells. Expression quantitative trait loci (eQTL) variants confirmed earlier DLEU2 associations. Furthermore, a novel variant (rs12585295) showed high localization with transcriptionally active chromatin states in osteoblast primary cell cultures. The supposition that DLEU2-encoded miRNAs have an important regulatory role in bone remodeling was further confirmed in a transgenic mice model showing that miR-15a/16-1-deleted mice had significantly higher percentage bone volume and trabecular number than the wild type, possibly due to prenatal actions. However, the three-point mechanical break force test of mice femurs showed a positive correlation between strength and miR-15a-5p/miR-16-5p levels, indicating differential effects on cortical and trabecular bone. Moreover, these miRNAs appear to have distinct and complex actions in mice prenatally and in adult humans, impacting BMD and microstructure by regulating bone cell transcription. However, detailed interactions between these miRNAs and their downstream mechanisms in health and disease need further clarification.
Collapse
Affiliation(s)
- Sjur Reppe
- Department of Medical Biochemistry, Oslo University Hospital, 0450 Oslo, Norway
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, 0456 Oslo, Norway
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Janne Elin Reseland
- Department of Biomaterials, Faculty of Dentistry, University of Oslo, 0317 Oslo, Norway
| | - Vid Prijatelj
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 Rotterdam, The Netherlands
| | - Michael Prediger
- Blood Sciences, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Newcastle upon Tyne NE2 4HH, UK
| | | | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, 0450 Oslo, Norway
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 Rotterdam, The Netherlands
| | - Kaare M Gautvik
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, 0456 Oslo, Norway
| | - Harish Kumar Datta
- Blood Sciences (Pathology), James Cook University Hospital, Middlesbrough TS4 3BW, UK
| |
Collapse
|
48
|
Shine BK, Choi JE, Park YJ, Hong KW. The Genetic Variants Influencing Hypertension Prevalence Based on the Risk of Insulin Resistance as Assessed Using the Metabolic Score for Insulin Resistance (METS-IR). Int J Mol Sci 2024; 25:12690. [PMID: 39684400 DOI: 10.3390/ijms252312690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Insulin resistance is a major indicator of cardiovascular diseases, including hypertension. The Metabolic Score for Insulin Resistance (METS-IR) offers a simplified and cost-effective way to evaluate insulin resistance. This study aimed to identify genetic variants associated with the prevalence of hypertension stratified by METS-IR score levels. Data from the Korean Genome and Epidemiology Study (KoGES) were analyzed. The METS-IR was calculated using the following formula: ln [(2 × fasting blood glucose (FBG) + triglycerides (TG)) × body mass index (BMI)]/ ln [high-density lipoprotein cholesterol (HDL-C)]. The participants were divided into tertiles 1 (T1) and 3 (T3) based on their METS-IR scores. Genome-wide association studies (GWAS) were performed for hypertensive cases and non-hypertensive controls within these tertile groups using logistic regression adjusted for age, sex, and lifestyle factors. Among the METS-IR tertile groups, 3517 of the 19,774 participants (17.8%) at T1 had hypertension, whereas 8653 of the 20,374 participants (42.5%) at T3 had hypertension. A total of 113 single-nucleotide polymorphisms (SNPs) reached the GWAS significance threshold (p < 5 × 10-8) in at least one tertile group, mapping to six distinct genetic loci. Notably, four loci, rs11899121 (chr2p24), rs7556898 (chr2q24.3), rs17249754 (ATP2B1), and rs1980854 (chr20p12.2), were significantly associated with hypertension in the high-METS-score group (T3). rs10857147 (FGF5) was significant in both the T1 and T3 groups, whereas rs671 (ALDH2) was significant only in the T1 group. The GWASs identified six genetic loci significantly associated with hypertension, with distinct patterns across METS-IR tertiles, highlighting the role of metabolic context in genetic susceptibility. These findings underscore critical genetic factors influencing hypertension prevalence and provide insights into the metabolic-genetic interplay underlying this condition.
Collapse
Affiliation(s)
- Bo-Kyung Shine
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Ja-Eun Choi
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| | - Young-Jin Park
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Kyung-Won Hong
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| |
Collapse
|
49
|
Zhang M, Yang E, Qin X, Zhang S, Zhu Y, Fu H, He B. EPSTI1 promotes osteoclast differentiation and bone resorption by PKR/NF-κB signaling. Biochem Biophys Res Commun 2024; 734:150463. [PMID: 39083969 DOI: 10.1016/j.bbrc.2024.150463] [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: 05/08/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Epithelial stromal interaction 1 (EPSTI1) plays an important role in M1 macrophages, which induce osteoclastogenesis. One recent genome-wide association study (GWAS) involving 426,824 individuals has shown that EPSTI1 is strongly associated with osteoporosis (P < 5E-8). Therefore, we speculate that EPSTI1 participates in the modulation of osteoporosis through osteoclastogenesis. The roles of EPSTI1 in osteoclastogenesis and bone resorption remain unclear. METHODS Femur specimens were collected from osteoporotic patients and control patients. Immunofluorescence staining was used to detect the expression of EPSTI1 and signaling pathways. The osteoclastic potential of RAW264.7 cells with Sh-EPSTI1 lentivirus infection was tested using tartrate-resistant acid phosphatase (TRAP) staining, western blotting, and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Western blotting was also used to examine signaling pathways. RESULTS In this study, EPSTI1 was found to be significantly increased in tartrate-resistant acid phosphatase positive (ACP5+) osteoclasts of bone sections from osteoporotic patients. Next, we identified EPSTI1 as a positive regulator of osteoclastogenesis and osteoclast differentiation capability. Diminished EPSTI1 expression resulted in reduced osteoclastic resorption. Mechanistically, EPSTI1-driven osteoclastogenesis was regulated by NF-κB pathway, which was mediated by the phosphorylation of protein kinase R (p-PKR). Furthermore, EPSTI1 participating in the modulation of osteoporosis via PKR/NF-κB pathway was also verified in the bone samples of osteoporotic patients. CONCLUSIONS Collectively, our findings suggest that EPSTI1 may regulate osteoclast differentiation and bone resorption through PKR/NF-κB pathway and in vivo experiments are needed to further verify EPSTI1 as the therapy target for osteoporosis.
Collapse
Affiliation(s)
- Muzi Zhang
- Department of Plastic Surgery, Medical Cosmetology Center of the First Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - E Yang
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Qin
- Department of Plastic Surgery, Medical Cosmetology Center of the First Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shun Zhang
- Department of Plastic Surgery, Medical Cosmetology Center of the First Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongyi Fu
- Department of Plastic Surgery, Medical Cosmetology Center of the First Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Bin He
- Department of Plastic Surgery, Medical Cosmetology Center of the First Branch, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
50
|
Coryell PR, Hardy PB, Chubinskaya S, Pearce KH, Loeser RF. A novel small molecule screening assay using normal human chondrocytes toward osteoarthritis drug discovery. PLoS One 2024; 19:e0308647. [PMID: 39485774 PMCID: PMC11530018 DOI: 10.1371/journal.pone.0308647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/26/2024] [Indexed: 11/03/2024] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis and a leading cause of pain and disability in adults. A central feature is progressive cartilage degradation and matrix fragment formation driven by the excessive production of matrix metalloproteinases (MMPs), such as MMP-13, by articular chondrocytes. Inflammatory factors, including interleukin 6 (IL-6), are secreted into the joint by synovial fibroblasts, and can contribute to pain and inflammation. No therapeutic exists that addresses the underlying loss of joint tissue in OA. To address this, we developed and utilized a cell-based high-throughput OA drug discovery platform using normal human chondrocytes treated with a recombinant fragment of the matrix protein fibronectin (FN-f) as a catabolic stimulus relevant to OA pathogenesis and a readout using a fluorescent MMP-13 responsive probe. The goal was to test this screening platform by identifying compounds that inhibited FN-f-induced MMP-13 production and determine if these compounds also inhibited catabolic signaling in OA chondrocytes and synovial fibroblasts. Two pilot screens of 1344 small molecules revealed five "hits" that strongly inhibited FN-f induced MMP-13 production with low cytotoxicity. These included RO-3306 (CDK1 inhibitor (i)), staurosporine (PKCi), trametinib (MEK1 and MEK2i), GSK-626616 (DYRK3i), and edicotinib (CSF-1Ri). Secondary testing using immunoblots and cells derived from OA joint tissues confirmed the ability of selected compounds to inhibit chondrocyte MMP-13 production and FN-f stimulated IL-6 production by synovial fibroblasts. These findings support the use of this high throughput screening assay for discovery of disease-modifying osteoarthritis drugs.
Collapse
Affiliation(s)
- Philip R. Coryell
- Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Paul B. Hardy
- Center for Integrative Chemical and Biological Drug Discovery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | - Kenneth H. Pearce
- Center for Integrative Chemical and Biological Drug Discovery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Richard F. Loeser
- Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|