1
|
Risi R, Amendolara R, Balena A, Watanabe M, Masi D, Fassino V, Luverà D, D'Onofrio L, Lauria A, Zampetti S, Gnessi L, Maddaloni E, Buzzetti R. Osteocalcin is inversely associated with worse adipose tissue distribution and cardiovascular risk in autoimmune diabetes. Diabetes Res Clin Pract 2025; 223:112114. [PMID: 40139321 DOI: 10.1016/j.diabres.2025.112114] [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: 11/15/2024] [Revised: 02/18/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Osteocalcin (OCN), whose release is impaired in diabetes, is suggested to regulate the adipose tissue (AT), being potentially associated with Cardiovascular risk (CVR). We aimed at evaluating whether OCN serum levels are associated with AT health and CVR in a primary CV prevention population with AD. METHODS Body mass composition was assessed in sixty-two people with AD.Serum levels of OCN, adipokines and markers of endothelial dysfunction were measured. Regression models were used to test the association of OCN with markers of AT, endothelial dysfunction and CVR categories as determined by the Steno Type 1 Risk Engine (ST1RE) score. RESULTS OCN was inversely associated with upper body fat deposition index (UBDFI) (Adj β coefficient -0.484, p value = 0.001). People in medium/high CV risk categories had higher UBFDI and lower OCN, while biomarkers of endothelial dysfunction were not different across CVR classes. A logistic binary regression for ST1RE score showed significant association of OCN with medium/high CVR category: OR [95 % CI for 1 SD increase: 0.541 [0.264--1.108], p = 0.093]. CONCLUSION OCN is inversely associated with unhealthy AT, supporting the protective role of OCN in AT. Moreover, lower OCN levels are associated with increased CVR in AD population.
Collapse
Affiliation(s)
- Renata Risi
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Rocco Amendolara
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Angela Balena
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Mikiko Watanabe
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Davide Masi
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Valeria Fassino
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Daniela Luverà
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Luca D'Onofrio
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Angelo Lauria
- Diabetology Unit, San Camillo Forlanini Hospital, Rome, Italy
| | - Simona Zampetti
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Ernesto Maddaloni
- Department of Experimental Medicine, Sapienza University of Rome, Italy.
| | | |
Collapse
|
2
|
He T, Qin L, Chen S, Huo S, Li J, Zhang F, Yi W, Mei Y, Xiao G. Bone-derived factors mediate crosstalk between skeletal and extra-skeletal organs. Bone Res 2025; 13:49. [PMID: 40307216 PMCID: PMC12044029 DOI: 10.1038/s41413-025-00424-1] [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: 10/28/2024] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Bone has long been acknowledged as a fundamental structural entity that provides support and protection to the body's organs. However, emerging research indicates that bone plays a crucial role in the regulation of systemic metabolism. This is achieved through the secretion of a variety of hormones, cytokines, metal ions, extracellular vesicles, and other proteins/peptides, collectively referred to as bone-derived factors (BDFs). BDFs act as a medium through which bones can exert targeted regulatory functions upon various organs, thereby underscoring the profound and concrete implications of bone in human physiology. Nevertheless, there remains a pressing need for further investigations to elucidate the underlying mechanisms that inform the effects of bone on other body systems. This review aims to summarize the current findings related to the roles of these significant modulators across different organs and metabolic contexts by regulating critical genes and signaling pathways in vivo. It also addresses their involvement in the pathogenesis of various diseases affecting the musculoskeletal system, circulatory system, glucose and lipid metabolism, central nervous system, urinary system, and reproductive system. The insights gained from this review may contribute to the development of innovative therapeutic strategies through a focused approach to bone secretomes. Continued research into BDFs is expected to enhance our understanding of bone as a multifunctional organ with diverse regulatory roles in human health.
Collapse
Affiliation(s)
- Tailin He
- Department of Rheumatology and Immunology, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), 100101, Beijing, China
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lei Qin
- Department of Orthopedics, Shenzhen Nanshan People's Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shaochuan Huo
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen 518000, China, Shenzhen Research Institute of Guangzhou University of Traditional Medicine (Futian), Shenzhen, 518000, China
| | - Jie Li
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Fuping Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), 100101, Beijing, China
| | - Weihong Yi
- Department of Orthopedics, Shenzhen Nanshan People's Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Yifang Mei
- Department of Rheumatology and Immunology, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
| | - Guozhi Xiao
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Boisen IM, Krarup Knudsen N, Nielsen JE, Kooij I, Bagger ML, Kaludjerovic J, O'Shaughnessy P, Andrews PW, Ide N, Toft BG, Juul A, Mehmedbasic A, Jørgensen A, Smith LB, Norman R, Meyts ERD, Lanske B, Blomberg Jensen M. Changes in local mineral homeostasis facilitate the formation of benign and malignant testicular microcalcifications. eLife 2025; 13:RP95545. [PMID: 40279260 PMCID: PMC12029210 DOI: 10.7554/elife.95545] [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: 04/27/2025] Open
Abstract
Testicular microcalcifications consist of hydroxyapatite and have been associated with an increased risk of testicular germ cell tumors (TGCTs) but are also found in benign cases such as loss-of-function variants in the phosphate transporter SLC34A2. Here, we show that fibroblast growth factor 23 (FGF23), a regulator of phosphate homeostasis, is expressed in testicular germ cell neoplasia in situ (GCNIS), embryonal carcinoma (EC), and human embryonic stem cells. FGF23 is not glycosylated in TGCTs and therefore cleaved into a C-terminal fragment which competitively antagonizes full-length FGF23. Here, Fgf23 knockout mice presented with marked calcifications in the epididymis, spermatogenic arrest, and focally germ cells expressing the osteoblast marker Osteocalcin (gene name: Bglap, protein name). Moreover, the frequent testicular microcalcifications in mice with no functional androgen receptor and lack of circulating gonadotropins are associated with lower Slc34a2 and higher Bglap/Slc34a1 (protein name: NPT2a) expression compared with wild-type mice. In accordance, human testicular specimens with microcalcifications also have lower SLC34A2 and a subpopulation of germ cells express phosphate transporter NPT2a, Osteocalcin, and RUNX2 highlighting aberrant local phosphate handling and expression of bone-specific proteins. Mineral disturbance in vitro using calcium or phosphate treatment induced deposition of calcium phosphate in a spermatogonial cell line and this effect was fully rescued by the mineralization inhibitor pyrophosphate. In conclusion, testicular microcalcifications arise secondary to local alterations in mineral homeostasis, which in combination with impaired Sertoli cell function and reduced levels of mineralization inhibitors due to high alkaline phosphatase activity in GCNIS and TGCTs facilitate osteogenic-like differentiation of testicular cells and deposition of hydroxyapatite.
Collapse
Affiliation(s)
- Ida Marie Boisen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of CopenhagenCopenhagen NDenmark
| | - Nadia Krarup Knudsen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - John E Nielsen
- Department of Growth and Reproduction, Rigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Ireen Kooij
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Mathilde Louise Bagger
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Jovanna Kaludjerovic
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Harvard UniversityBostonUnited States
| | - Peter O'Shaughnessy
- School of Biodiversity, One Health & Veterinary Medicine, University of GlasgowGlasgowUnited Kingdom
| | - Peter W Andrews
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Western BankSheffieldUnited Kingdom
| | - Noriko Ide
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Harvard UniversityBostonUnited States
| | | | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Arnela Mehmedbasic
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Anne Jørgensen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research InstituteEdinburghUnited Kingdom
| | - Richard Norman
- Department of Urology, Dalhousie UniversityHalifaxCanada
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Rigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Beate Lanske
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Harvard UniversityBostonUnited States
| | - Martin Blomberg Jensen
- Division of Translational Endocrinology, Department of Endocrinology and Internal Medicine, University Hospital Copenhagen, Herlev-GentofteHerlevDenmark
- Department of Clinical Medicine, Copenhagen University HospitalCopenhagenDenmark
| |
Collapse
|
4
|
Bighetti-Trevisan RL, Almeida LO, Ramos JIR, Freitas GP, Oliveira FS, Gordon JAR, Tye CE, Stein GS, Lian JB, Stein JL, Rosa AL, Beloti MM. The effect of osteoclasts on epigenetic regulation by long non-coding RNAs in osteoblasts grown on titanium with nanotopography. BIOMATERIALS ADVANCES 2025; 168:214128. [PMID: 39622096 DOI: 10.1016/j.bioadv.2024.214128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/05/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024]
Abstract
Titanium (Ti) implant osseointegration is regulated by the crosstalk among bone cells that are affected by epigenetic machinery, including the regulation of long non-coding RNAs (lncRNAs). Nanotopography Ti (Ti Nano) induces the differentiation of osteoblasts that are inhibited by osteoclasts through epigenetic mechanisms. Thus, we hypothesize that osteoclasts affect lncRNA expression in Ti Nano-cultivated osteoblasts. Osteoblasts were grown on Ti Nano and Ti Control that were then co-cultured with osteoclasts for 48 h. Using RNAseq, we identified 252 modulated lncRNAs in osteoblasts regulated by both surfaces of Ti, but mainly in Ti Nano-cultivated osteoblasts. A negative correlation was observed between Kcnq1ot1 and the mRNAs of Alpl, Bglap, Bmp8a, Col1a1, and Vim in Ti Nano-cultivated osteoblasts with osteoclasts. The pull-down indicated that Bglap mRNA is a direct target of Kcnq1ot1, with enhanced physical interaction in Ti Nano-cultivated osteoblasts, and greater osteoclast inhibition than the Ti Control. The bone marker expression at the levels of mRNA and protein were downregulated by the Kcnq1ot1 silencing, indicating its pivotal role in osteoblast differentiation. These results showed that nanostructured Ti surface modulates the osteoblast-osteoclast crosstalk, at least in part, through the regulation of lncRNA expression in osteoblasts. We demonstrate that the lncRNA Kcnq1ot1 directly interacts with Bglap mRNA, and this interaction is enhanced by nanotopography and reduced by osteoclasts with greater intensity in Ti Nano-cultivated osteoblasts. These findings confirm the molecular mechanisms associated with the high osteogenic potential of nanotopography and can potentially support osteointegration of dental and skeletal prostheses.
Collapse
Affiliation(s)
- Rayana Longo Bighetti-Trevisan
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil; Faculty of Dentistry, University of Ribeirão Preto, Ribeirão Preto, SP, Brazil.
| | - Luciana Oliveira Almeida
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil; Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | | | - Gileade Pereira Freitas
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil; Department of Oral and Maxillofacial Surgery, School of Dentistry, Federal University of Goiás, Goiânia, GO, Brazil..
| | | | - Jonathan Alexander Robert Gordon
- Department of Biochemistry and Vermont Cancer Center, Larner College of Medicine, University of Vermont, Burlington, VT, United States of America.
| | - Coralee Elizabeth Tye
- Department of Biochemistry and Vermont Cancer Center, Larner College of Medicine, University of Vermont, Burlington, VT, United States of America.
| | - Gary Stephen Stein
- Department of Biochemistry and Vermont Cancer Center, Larner College of Medicine, University of Vermont, Burlington, VT, United States of America.
| | - Jane Barbara Lian
- Department of Biochemistry and Vermont Cancer Center, Larner College of Medicine, University of Vermont, Burlington, VT, United States of America.
| | - Janet Lee Stein
- Department of Biochemistry and Vermont Cancer Center, Larner College of Medicine, University of Vermont, Burlington, VT, United States of America.
| | - Adalberto Luiz Rosa
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Marcio Mateus Beloti
- Bone Research Lab, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
5
|
Kirk B, Lombardi G, Duque G. Bone and muscle crosstalk in ageing and disease. Nat Rev Endocrinol 2025:10.1038/s41574-025-01088-x. [PMID: 40011751 DOI: 10.1038/s41574-025-01088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
Interorgan communication between bone and skeletal muscle is central to human health. A dysregulation of bone-muscle crosstalk is implicated in several age-related diseases. Ageing-associated changes in endocrine, inflammatory, nutritional and biomechanical stimuli can influence the differentiation capacity, function and survival of mesenchymal stem cells and bone-forming and muscle-forming cells. Consequently, the secretome phenotype of bone and muscle cells is altered, leading to impaired crosstalk and, ultimately, catabolism of both tissues. Adipose tissue acts as a third player in the bone-muscle interaction by secreting factors that affect bone and muscle cells. Physical exercise remains the key biological stimulus for bone-muscle crosstalk, either directly via the release of cytokines from bone, muscle or adipocytes, or indirectly through extracellular vesicles. Overall, bone-muscle crosstalk is considered an inherent process necessary to maintain the structure and function of both tissues across the life cycle. This Review summarizes the latest biomedical advances in bone-muscle crosstalk as it pertains to human ageing and disease. We also outline future research priorities to accommodate the understanding of this rapidly emerging field.
Collapse
Affiliation(s)
- Ben Kirk
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Gustavo Duque
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia.
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
6
|
Guo X, Hou C, Liu F, Zhou R, Tian G, Liu JM, Li R. Genetic insights into circulating osteocalcin for cardiovascular diseases and the role of vascular calcification. Nutr Metab Cardiovasc Dis 2025:103870. [PMID: 39986934 DOI: 10.1016/j.numecd.2025.103870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 12/04/2024] [Accepted: 01/22/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND AND AIMS Studies have suggested that osteocalcin (OCN) is implicated in vascular calcification and linked to cardiovascular diseases (CVDs), but it is unclear whether the relationships are causal. The aim of this study is to evaluate the causal relationship of circulating OCN with CVDs and the role of vascular calcification. METHODS AND RESULTS Bi-directional, mediation, and multivariable Mendelian randomization (MVMR) were performed using summary-level data for circulating OCN levels, coronary artery calcification (CAC), and CVDs, including coronary artery disease (CAD), myocardial infarction (MI), heart failure, atrial fibrillation, stroke and its subtypes. Pooled estimates from two independent datasets of OCN were calculated using the inverse variance weighted method with sensitivity analyses. The conservative Hochberg correction method adjusted the P-value for multiple comparisons. Genetically predicted higher OCN levels were linked to an increased risk of CAD (odds ratio [OR] = 1.069, 95%CI = 1.037-1.102, P < 0.001) and MI (OR = 1.099, 95%CI = 1.069-1.130, P < 0.001). In addition, elevated OCN levels were associated with higher CAC (β = 0.180, 95%CI = 0.101-0.258, P = 0.006), which was related higher risk of CAD (OR = 1.225, 95%CI = 1.132-1.325, P < 0.001) and MI (OR = 1.286, 95%CI = 1.203-1.375, P < 0.001), mediating 54.5 % and 48.3 % of the effect of OCN on CAD and MI, respectively. Meanwhile, MVMR results also validated the mediating role of CAC. In contrast, CAD and MI were associated with decreased levels of plasma OCN. CONCLUSION Our findings reveal that higher OCN concentrations are associated with an elevated risk of CAD and MI, which was partially mediated by CAC. Lower OCN levels found in previous observational studies might be due to reverse causation.
Collapse
Affiliation(s)
- Xingzhi Guo
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China; Department of Geriatric Neurology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, China
| | - Chen Hou
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China; Department of Geriatric Neurology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Fuqiang Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, 710068, Shaanxi, China
| | - Rong Zhou
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China; Department of Geriatric Neurology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, China
| | - Ge Tian
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China; Department of Geriatric Neurology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Rui Li
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China; Department of Geriatric Neurology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China; Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, China.
| |
Collapse
|
7
|
Liu Z, Mao Y, Yang K, Wang S, Zou F. A trend of osteocalcin in diabetes mellitus research: bibliometric and visualization analysis. Front Endocrinol (Lausanne) 2025; 15:1475214. [PMID: 39872315 PMCID: PMC11769813 DOI: 10.3389/fendo.2024.1475214] [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: 08/03/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
Background Osteocalcin has attracted attention for its potential role in diabetes management. However, there has been no bibliometric assessment of scientific progress in this field. Methods We analysed 1680 articles retrieved from the Web of Science Core Collection (WoSCC) between 1 January 1986 and 10 May 2024 using various online tools. Result These papers accumulated 42,714 citations,with an average of 25.43 citations per paper. Publication output increased sharply from 1991 onwards. The United States and China are at the forefront of this research area. Discussion The keywords were grouped into four clusters: 'Differential and functional osteocalcin genes', 'Differential expression of osteocalcin genes in relation to diabetes mellitus', 'Role of osteocalcin in the assessment of osteoporosis and diabetes mellitus', and 'Indirect involvement of osteocalcin in metabolic processes'. Analysis using the VoS viewer suggests a shift in research focus towards the correlation between osteocalcin levels and diabetic complications, the clinical efficacy of therapeutic agents or vitamins in the treatment of osteoporosis in diabetic patients, and the mechanisms by which osteocalcin modulates insulin action. The proposed focus areas are "osteocalcin genes", "insulin regulation and osteoporosis ", "different populations", "diabetes-related complications" and "type 2 diabetes mellitus","effect of osteocalcin expression on insulin sensitivity as well as secretion","osteocalcin expression in different populations of diabetic patients and treatment-related studies".
Collapse
Affiliation(s)
- Zixu Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Yuchen Mao
- The First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Kangping Yang
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Shukai Wang
- The First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Fang Zou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Tong X, Fu X, Gong A, Yu G, Chen N, Chen B, Gu J, Liu Z. Effect of Luteolin on cadmium-inhibited bone growth via suppressing osteoclastogenesis in laying chickens. J Anim Sci 2025; 103:skaf033. [PMID: 39921628 PMCID: PMC11912829 DOI: 10.1093/jas/skaf033] [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: 11/18/2024] [Accepted: 02/07/2025] [Indexed: 02/10/2025] Open
Abstract
Luteolin (Lut) is a flavonoid derived from several plant sources. Cadmium (Cd) is a widespread environmental contaminant and potential toxin with detrimental effects on animal health. However, the effect of Lut on Cd-induced inhibition of bone growth in laying chickens remains unclear. This study investigates the effects of Lut on Cd-induced inhibition of bone growth in the femur and tibia of laying chickens. A total of sixty 1-d-old green-eggshell yellow feather laying chickens were randomly assigned to 4 groups after a 5-d acclimation period: basal diet (Con), cadmium chloride (CdCl2, Cd), Lut, and Lut + Cd. Bone microstructure, serum biomarkers of bone remodeling, the levels of Cd, calcium (Ca), phosphorus (P), and trace metal elements were assessed using the micro-computed tomography (Micro-CT), enzyme-linked immunosorbent assay (ELISA), and microwave digestion, respectively. Bone remodeling biomarkers, late endosomal/lysosomal adaptor and MAPK and mTOR activator 1 (LAMTOR1), as well as the phosphorylation of AMP-activated protein kinase α (AMPKα) and protein kinase B (Akt), were quantified using the qRT-PCR and western blot. The results indicated that Lut effectively mitigated Cd-induced bone mass loss compared to the Cd group, resulting in increased bone volume (BV), bone surface/BV (BS/BV), connectivity density (Conn.Dn), and the length and weight of the femur and tibia in laying chickens. Mechanistically, compared to the Cd group, Lut restored the ratio of osteoprotegerin (OPG)/receptor activator of NF-κB ligand (RANKL) in serum and bone tissue, enhanced the expression of bone morphogenetic protein-2 (BMP-2), runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and Osterix (OSX), while reducing the levels of Ca, Cd, and alkaline phosphatase (ALP) activity, as well as the expression of osteopontin (OPN), c-Fos, osteoclast stimulatory-transmembrane protein (OC-STAMP), tartrate-resistant acid phosphatase, cathepsin K (CTSK), matrix metalloprotein-9 (MMP-9), LAMTOR1, and the phosphorylation of AMPKα and Akt. Therefore, Lut alleviates Cd-induced damage to the femur and tibia of chickens by promoting osteogenesis and inhibiting osteoclastogenesis, positioning Lut as a potential therapeutic plant extract for enhancing bone growth in laying chickens.
Collapse
Affiliation(s)
- Xishuai Tong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Xiaohui Fu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Anqing Gong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Gengsheng Yu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Naineng Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Bing Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Jianhong Gu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Zongping Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| |
Collapse
|
9
|
Jin Z, Chen Z, Liang T, Liu W, Shan Z, Tan D, Chen J, Hu J, Qin L, Xu J. Accelerated fracture healing accompanied with traumatic brain injury: A review of clinical studies, animal models and potential mechanisms. J Orthop Translat 2025; 50:71-84. [PMID: 39868349 PMCID: PMC11763218 DOI: 10.1016/j.jot.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 01/28/2025] Open
Abstract
The orthopaedic community frequently encounters polytrauma individuals with concomitant traumatic brain injury (TBI) and their fractures demonstrate accelerated fracture union, but the mechanisms remain far from clear. Animal and clinical studies demonstrate robust callus formation at the early healing process and expedited radiographical union. In humans, robust callus formation in TBI occurs independently of fracture fixation methods across multiple fracture sites. Animal studies of TBI replicate clinically relevant enlarged fracture callus as characterized by increased tissue volume and bone volume at the early stages. However, refinement and standardization of the TBI models requires further research. The quest for its underlying mechanisms began with the finding of increased osteogenesis in vitro using the serum and cerebral spinal fluid (CSF) from TBI individuals. This has led to the investigation of myriads of brain-derived factors including humoral factors, cytokines, exosomes, and mi-RNAs. Further, the emerging information of interplay between the skeletal system and central nervous system, the roles of peripheral nerves and their neuropeptides in regulating bone regeneration, offers valuable insights for future research. This review consolidates the findings from both experimental and clinical studies, elucidating the potential mechanisms underlying enhanced fracture healing in concurrent TBI scenarios that may lay down a foundation to develop innovative therapies for fracture healing enhancement and conquer fracture non-union. The translational potential of this article. This review comprehensively summarizes the observations of accelerated fracture healing in the presence of traumatic brain injury from both preclinical and clinical studies. In addition, it also delineates potential cellular and molecular mechanisms. Further detailed investigation into its underlying mechanisms may reveal innovative orthopaedic intervention strategies to improve fracture healing and thus offering promising avenues for future translational applications.
Collapse
Affiliation(s)
- Zheyu Jin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Tongzhou Liang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Weiyang Liu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhengming Shan
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dianhui Tan
- Department of Neurosurgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiechen Chen
- Department of Orthopaedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Orthopaedic Medical Research Centre, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jun Hu
- Department of Orthopaedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Orthopaedic Medical Research Centre, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Areas of Excellence Centre for Musculoskeletal Degeneration and Regeneration, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
10
|
Taib IS, Jayusman PA. The Role of Bone-Derived Osteocalcin in Testicular Steroidogenesis: Contributing Factor to Male Fertility. Diseases 2024; 12:335. [PMID: 39727665 PMCID: PMC11727589 DOI: 10.3390/diseases12120335] [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: 10/11/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Osteocalcin (OCN), a protein predominantly produced by osteoblasts in bone, has emerged as a significant factor in bone metabolism and reproductive function. This article reviews the latest research on the role of OCN beyond its traditional functions in bone mineralisation, particularly its influence on testicular steroidogenesis and male fertility. The structure and modifications of OCN are elaborated upon, highlighting its uncarboxylated form (ucOCN), which is becoming increasingly recognised for its bioactive properties. The impact of OCN on bone quantity, quality and strength is summarised, emphasising its role as a regulator of bone metabolism. Furthermore, the influence of ucOCN on testicular steroidogenesis and the involvement of GPRC6A, a G protein-coupled receptor, in mediating these effects are also explored. Evidence suggests that ucOCN regulates testosterone synthesis and spermatogenesis, which indirectly have the potential to influence bone metabolism integrity. In conclusion, OCN, particularly in its uncarboxylated form, plays a crucial role in bone metabolism and male fertility by regulating testicular steroidogenesis, with GPRC6A mediating these effects, thereby linking bone health and reproductive functions.
Collapse
Affiliation(s)
- Izatus Shima Taib
- Centre of Diagnostics, Therapeutics and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Putri Ayu Jayusman
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
11
|
Aldogan EH, Başaran D, Öner B, Günçer B. The Role of Formononetin in Osteoblast Function and Mineralization Potential with Deproteinized Bovine Bone Material. Curr Issues Mol Biol 2024; 46:14215-14225. [PMID: 39727979 DOI: 10.3390/cimb46120851] [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: 10/15/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES Dental bone formation involves various cellular and molecular mechanisms, and phytoestrogens such as formononetin (FORM) are promising because of their estrogenic, anti-inflammatory, and antioxidant effects. This study investigated the effect of FORM on osteoblast proliferation, differentiation, and mineralization in combination with spongiosa granulates (BO) in vitro. MATERIALS AND METHODS Human fetal osteoblast cells (hFOB1.19) were treated with increasing concentrations of FORM (1, 10, and 100 µg/mL), BO, or their combination. Cell proliferation was assessed using a MTT assay. Alkaline phosphatase (ALP) activity, intracellular Ca2+, and Pi levels were measured using ELISA. Vascular endothelial growth factor (VEGF) and osteocalcin expression levels were analyzed by western blotting. RESULTS Cell proliferation increased with FORM, with or without BO, after 6 days (p < 0.001). FORM and BO had a synergistic effect on ALP activity (p < 0.001). Intracellular Ca2+ and Pi levels were highest in the BO-FORM group, suggesting superior mineralization (p < 0.05). VEGF and osteocalcin expression was significantly upregulated with FORM, alone and with BO (p < 0.05), indicating improved angiogenesis and bone maturation over 9 days. CONCLUSIONS FORM enhances osteoblast proliferation, differentiation, and mineralization potential, particularly in BO spongiosa granulates. These data support the in vitro potential of formononetin-phytoestrogen in promoting osteoblast differentiation and mineralization potential with BO. These findings suggest that FORM, combined with BO, could improve bone augmentation in clinical applications such as maxillofacial surgery. FORM shows valuable potential for clinical applications, such as maxillofacial surgery, by promoting faster and more effective healing.
Collapse
Affiliation(s)
- Ebru Haciosmanoglu Aldogan
- Department of Biophysics, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, 34098 Fatih, Istanbul, Turkey
| | - Deniz Başaran
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Istanbul University, 34093 Fatih, Istanbul, Turkey
| | - Bilgin Öner
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Istanbul University, 34093 Fatih, Istanbul, Turkey
| | - Başak Günçer
- Department of Biophysics, Istanbul Faculty of Medicine, Istanbul University, 34093 Fatih, Istanbul, Turkey
| |
Collapse
|
12
|
Sartori G, Bertoldo F, Gretter A, Lovati FM, Caprino R, Viterale G, Crisafulli E. Impact of the visceral adipose tissue on bone quality in patients with untreated mild-to-severe obstructive sleep apnea. J Sleep Res 2024:e14397. [PMID: 39658313 DOI: 10.1111/jsr.14397] [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: 09/16/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024]
Abstract
Obstructive sleep apnea (OSA) predominantly affects patients who are obese and causes systemic organ damage. Little is known about the relationship between fat distribution and bone impairment in these patients. We aimed to evaluate the impact of the visceral adipose tissue (VAT) on the bone quality of patients with OSA. In our prospective study, 49 untreated patients with mild-to-severe OSA underwent dual-energy X-ray absorptiometry. Polygraphy data were also collected. According to the recent reference values for European adults, patients were divided by the sex-related threshold of the VAT index into two categories: VAT index within limits (normal VAT [nVAT]) and increased VAT (iVAT). In all, 63% of the patients were in the iVAT category. Compared to patients with nVAT, those with iVAT had a higher prevalence of arterial hypertension (52% versus 22%) and diabetes (32% versus 6%), and higher values of mean nocturnal desaturation. Patients with iVAT had, in comparison to those with nVAT, lower values of the lumbar spine trabecular bone score (TBS; mean 1.24 versus 1.39; p < 0.001), TBS T-score (mean -1.82 versus -0.52; p < 0.001) and TBS Z-score (mean -0.35 versus 0.75; p = 0.002). Moreover, a close association was present between the VAT index and TBS lumbar spine L1-L4 (r2 linear 0.573; p < 0.001), and altered values of the TBS Z-score were associated with the severity of vertebral fractures. Finally, in a linear regression-adjusted model, the VAT index predicted TBS lumbar spine L1-L4 (β -0.323; p < 0.001). In patients with OSA VAT impacts bone quality. In these patients, the role of VAT as a metabolically active tissue should be considered.
Collapse
Affiliation(s)
- Giulia Sartori
- Department of Medicine, Respiratory Medicine Unit, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Francesco Bertoldo
- Emergency Medicine Unit, Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Andrea Gretter
- Department of Medicine, Respiratory Medicine Unit, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Federica Margherita Lovati
- Department of Medicine, Respiratory Medicine Unit, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Rosaria Caprino
- Department of Medicine, Respiratory Medicine Unit, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Giovanni Viterale
- Department of Medicine, Respiratory Medicine Unit, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Ernesto Crisafulli
- Department of Medicine, Respiratory Medicine Unit, University of Verona and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| |
Collapse
|
13
|
Kim TH, Kim H, Lee HH, Sang JH. Vitamin K: Calcium Metabolism Modulator for Menopausal Women. J Menopausal Med 2024; 30:152-163. [PMID: 39829192 PMCID: PMC11745727 DOI: 10.6118/jmm.24023] [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: 08/18/2024] [Revised: 10/13/2024] [Accepted: 11/11/2024] [Indexed: 01/22/2025] Open
Abstract
Vitamin K (VitK) exists in multiple forms, with Vitamin K1 (VitK1) and Vitamin K2 (VitK2) being the most prominent. VitK1 primarily regulates clotting factors in the liver, whereas VitK2 plays a crucial role in activating extrahepatic proteins involved in various physiological processes. VitK plays a pivotal role in various physiological functions, including vascular health, bone metabolism, neuroprotection, hepatoprotection, immune response modulation, dental health, and glucose control. Particularly, activation of the matrix Gla protein and osteocalcin through VitK2 inhibits vascular calcification (VC) and promotes bone mineralization. This review provides an overview of the physiological functions of VitK2, underscoring its role in calcium metabolism modulation and its diverse effects on health. Additionally, this article provides a comprehensive overview of the beneficial functions of VitK, and discusses the significance of adequate dietary intake and oral supplementation of VitK. Particularly, emphasizing on the need for VitK2 supplementation owing to its relatively limited availability in Western diets. VitK2 supplementation effectively counters VC, enhances bone density, and offers neuroprotective, hepatoprotective, and anti-inflammatory benefits. Thus, the supplementation of VitK2, alongside dietary intake, is essential for preventive healthcare, particularly in the prevention of osteoporosis and vascular diseases. Incorporating adequate VitK2 intake highlights its significance in promoting overall well-being. Illustrated summary of the role of VitK in menopausal women.
Collapse
Affiliation(s)
- Tae-Hee Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Hayeon Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Hae Hyeog Lee
- Department of Obstetrics and Gynecology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea.
| | - Jae Hong Sang
- Department of Obstetrics and Gynecology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea.
| |
Collapse
|
14
|
Bernhard M, Okorie O, Tseng WJ, Chen M, Danon J, Cui M, Lashbrooks E, Yang Y, Wang B. Metabolic shifts in ratio of ucOcn to cOcn toward bone resorption contribute to age-dependent bone loss in male mice. Am J Physiol Endocrinol Metab 2024; 327:E711-E722. [PMID: 39441240 PMCID: PMC11684868 DOI: 10.1152/ajpendo.00294.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
The study of the senile osteoporosis in men still lags significantly behind that in women. The changes of protein molecule levels and their relationships with bone loss remain poorly understood. In the present study, we used C57BL/6J male mice at ages from 3 to 24 mo to delineate the mechanisms of aging effects on bone loss. We used the microcomputed tomography, mechanical testing, histomorphometry assays, and detection of serum levels of undercarboxylated osteocalcin (ucOcn) and carboxylated osteocalcin (cOcn) to assess bone mass changes and their relationships with the ratios of ucOcn-to-cOcn in mice from different age groups. The results showed that mouse trabecular bone mass reduced gradually with age, whereas cortical bone loss and mechanical property changes mostly occurred in advanced age. Our findings further demonstrated that the increase in osteoclast activity and the decrease in osteoblast function were significantly corelated with blood levels of ucOcn and cOcn, respectively. The dynamic metabolic changes of ucOcn to cOcn ratio were correlated with age-dependent bone loss in mice. In summary, metabolic shifts in the ratio of ucOcn to cOcn toward bone resorption from young adult to elderly mice contribute to the pathogenesis of age-related bone loss. Simultaneously monitoring blood ratios of ucOcn-to-cOcn may be useful to predict the status of bone mass in vivo.NEW & NOTEWORTHY To our knowledge, our finding in this study shows for the first time that metabolic shifts in ratio of ucOcn to cOcn toward bone resorption are markedly correlated with age-dependent bone loss in male mice. These findings for the effects of aging on bone loss will assist in studying the pathogenesis of human type II osteoporosis.
Collapse
Affiliation(s)
- Matthew Bernhard
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Obinna Okorie
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Wei-Ju Tseng
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Mengcun Chen
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Julia Danon
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Mingshu Cui
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Elisabeth Lashbrooks
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Yanmei Yang
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Bin Wang
- Departments of Medicine, The Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
15
|
Kalev-Altman R, Fraggi-Rankis V, Monsonego-Ornan E, Sela-Donenfeld D. The bone Gla protein osteocalcin is expressed in cranial neural crest cells. BMC Res Notes 2024; 17:329. [PMID: 39501347 PMCID: PMC11539830 DOI: 10.1186/s13104-024-06990-7] [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: 06/20/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Osteocalcin is a small protein abundant in the bone extracellular-matrix, that serves as a marker for mature osteoblasts. To become activated, osteocalcin undergoes a specific post-translational carboxylation. Osteocalcin is expressed at advanced stages of embryogenesis and after birth, when bone formation takes place. Neural crest cells (NCCs) are a unique cell population that evolves during early stages of development. While initially NCCs populate the dorsal neural-tube, later they undergo epithelial-to-mesenchymal-transition and migrate throughout the embryo in highly-regulated manner. NCCs give rise to multiple cell types including neurons and glia of the peripheral nervous system, chromaffin cells and skin melanocytes. Remarkably, in the head region, NCCs give rise to cartilage and bone. FINDING Here we report that osteocalcin is detected in cranial NCCs. Analysis of chick embryos at stages of cranial NCC migration revealed that osteocalcin mRNA and protein is expressed in pre-migratory and migratory NCCs in-vivo and ex-vivo. Addition of warfarin, an inhibitor of osteocalcin carboxylation, onto neural-tube explants, reduced the amount of NCC migration. These results provide the first evidence of osteocalcin presence in cranial NCCs, much before they give rise to craniofacial skeleton, and propose its possible involvement in the regulation of NCC migration.
Collapse
Affiliation(s)
- Rotem Kalev-Altman
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- The Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- The Section of Genetic Medicine, The Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Veatriki Fraggi-Rankis
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- Smart Assays Biotechnologies Ltd, Weizmann Science Park, Ness Ziona, 7414003, Israel
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel.
| |
Collapse
|
16
|
Jang HJ, Kang MS, Jang J, Lim D, Choi SW, Jung TG, Chun HJ, Kim B, Han DW. Harnessing 3D printed highly porous Ti-6Al-4V scaffolds coated with graphene oxide to promote osteogenesis. Biomater Sci 2024; 12:5491-5503. [PMID: 39310945 DOI: 10.1039/d4bm00970c] [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: 10/23/2024]
Abstract
Bone tissue engineering (BTE) strategies have been developed to address challenges in orthopedic and dental therapy by expediting osseointegration and new bone formation. In this study, we developed irregular porous Ti-6Al-4V scaffolds coated with reduced graphene oxide (rGO), specifically rGO-pTi, and investigated their ability to stimulate osseointegration in vivo. The rGO-pTi scaffolds exhibited unique irregular micropores and high hydrophilicity, facilitating protein adsorption and cell growth. In vitro assays revealed that the rGO-pTi scaffolds increased alkaline phosphatase (ALP) activity, mineralization nodule formation, and osteogenic gene upregulation in MC3T3-E1 preosteoblasts. Moreover, in vivo transplantation of rGO-pTi scaffolds in rabbit calvarial bone defects showed improved bone matrix formation and osseointegration without hemorrhage. These findings highlight the potential of combining rGO with irregular micropores as a promising BTE scaffold for bone regeneration.
Collapse
Affiliation(s)
- Hee Jeong Jang
- Department of Cogno-mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea.
| | - Moon Sung Kang
- Department of Cogno-mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea.
| | - Jinju Jang
- Department of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Corporate Research Institute, RNX Inc., Bucheon 14558, Republic of Korea
| | - Dohyung Lim
- Corporate Research Institute, RNX Inc., Bucheon 14558, Republic of Korea
- Department of Mechanical Engineering, Sejong University, Seoul 05006, Republic of Korea
| | - Seong-Won Choi
- Industry Support Center for Convergence Medical Devices, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Tae-Gon Jung
- Medical Device Development Center, Osong Medical Innovation Foundation, Chungju 28160, Republic of Korea
| | - Heoung-Jae Chun
- Department of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Bongju Kim
- Dental Life Science Research Institute/Innovation Research & Support Center for Dental Science, Seoul National University Dental Hospital, Seoul 03080, Republic of Korea
| | - Dong-Wook Han
- Department of Cogno-mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea.
- BIO-IT Fusion Technology Research Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
17
|
Meslier QA, Duerr TJ, Guan W, Nguyen B, Monaghan JR, Shefelbine SJ. WISH-BONE: Whole-mount in situ histology, to label osteocyte mRNA and protein in 3D adult mouse bones. FASEB J 2024; 38:e70101. [PMID: 39387181 DOI: 10.1096/fj.202400635r] [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: 03/22/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Bone is a three-dimensional (3D) highly dynamic tissue under constant remodeling. Commonly used tools to investigate bone biology require sample digestion for biomolecule extraction or provide only two-dimensional (2D) spatial information. There is a need for 3D tools to investigate spatially preserved biomarker expression in osteocytes. In this work, we present a new method, WISH-BONE, to label osteocyte messenger RNA (mRNA) and protein in whole-mount mouse bone. For mRNA labeling, we used hybridization chain reaction-fluorescence in situ hybridization (HCR-FISH) to label genes of interest in osteocytes. For protein labeling, samples were preserved using an epoxy-based solution that protects tissue structure and biomolecular components. Then an enzymatic matrix permeabilization step was performed to enable antibody penetration. Immunostaining was used to label various proteins involved in bone homeostasis. We also demonstrate the use of customized fluorescent nanobodies to target and label proteins in the cortical bone (CB). However, the relatively dim signal observed from nanobodies' staining limited detection. mRNA and protein labeling were performed in separate samples. In this study, we share protocols, highlight opportunities, and identify the challenges of this novel 3D labeling method. They are the first protocols for whole-mount osteocyte 3D labeling of mRNA and protein in mature mouse bones. WISH-BONE will allow the investigation of molecular signaling in bone cells in their 3D environment and could be applied to various bone-related fields of research.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - Timothy J Duerr
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| | - Webster Guan
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - Brian Nguyen
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - James R Monaghan
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Du X, Guo S, Mu X, Mei S, Yang R, Zhang H, Jiang C, Zhang J. Bencaosome [16:0 Lyso PA+XLGB28-sRNA] improves osteoporosis by simultaneously promoting osteogenesis and inhibiting osteoclastogenesis in mice. IUBMB Life 2024; 76:832-844. [PMID: 39012196 DOI: 10.1002/iub.2857] [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: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 07/17/2024]
Abstract
Osteoporosis (OP) is a systemic metabolic bone disease resulting in reduced bone strength and increased susceptibility to fractures, making it a significant public health and economic problem worldwide. The clinical use of anti-osteoporosis agents is limited because of their serious side effects or the high cost of long-term use. The Xianlinggubao (XLGB) formula is an effective traditional Chinese herbal medicine commonly used in orthopedics to treat osteoporosis; however, its mechanism of action remains unclear. In this study, we screened 40 small RNAs derived from XLGB capsules and found that XLGB28-sRNA targeting TNFSF11 exerted a significant anti-osteoporosis effect in vitro and in vivo by simultaneously promoting osteogenesis and inhibiting osteoclastogenesis. Oral administration of bencaosome [16:0 Lyso PA+XLGB28-sRNA] effectively improved bone mineral density and reduced the damage to the bone microstructure in mice. These results suggest that XLGB28-sRNA may be a novel oligonucleotide drug that promotes osteogenesis and inhibits osteoclastogenesis in mice.
Collapse
Affiliation(s)
- Xinyi Du
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shaoting Guo
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xuemeng Mu
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Song Mei
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Rui Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Hengyan Zhang
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Chengyu Jiang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jia Zhang
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| |
Collapse
|
19
|
He S, Hu R, Yao X, Cui J, Liu H, Zhu M, Ning L. The effects of heat and hydrogen peroxide treatment on the osteoinductivity of demineralized cortical bone: a potential method for preparing tendon/ligament repair scaffolds. Regen Biomater 2024; 11:rbae116. [PMID: 39398284 PMCID: PMC11471265 DOI: 10.1093/rb/rbae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/07/2024] [Indexed: 10/15/2024] Open
Abstract
Recent studies have indicated that demineralized cortical bone (DCB) may be used to repair tendons and ligaments, such as the patellar tendon and anterior cruciate ligament (ACL). Hydrogen peroxide (H2O2) has been shown to reduce the osteoinductivity of DCB, and heat treatment may also decrease the osteoinductivity of DCB. The purpose of this study was (i) to determine whether heat treatment reduces the osteoinductivity of DCB and (ii) to compare the effectiveness of heat treatment and H2O2 treatment on BMP-2 inactivation. DCB was prepared by immersion in 0.6 N hydrochloric acid, and DCB-H and DCB-HO were prepared by heat treatment (70°C for 8 h) and H2O2 treatment (3% H2O2 for 8 h), respectively. The surface topographies, elemental distributions and histological structures of the scaffolds were observed by scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FT-IR) and histological staining. The viability and osteogenic differentiation of TDSCs cultured on the scaffolds were evaluated via live/dead cell staining and Cell Counting Kit-8 (CCK-8) testing, real-time polymerase chain reaction (RT-PCR) and western bolt (WB) analysis, alkaline phosphatase activity (ALP) and alizarin red S (ARS) staining. The intramuscular implantation of the scaffolds in rats was also used to evaluate the effect of heat treatment and H2O2 treatment on the osteoinductivity of DCB. Our results demonstrated that both treatments removed BMP-2 and osteocalcin (OCN) within the DCB and that DCB-H and DCB-HO had good cytocompatibility and reduced the osteogenic differentiation of TDSCs. Moreover, the in vivo results indicated that the DCB-H and DCB-HO groups had smaller areas of osteoid formation than did the DCB group, and the DCB-HO group had the smallest area among the three groups. Our study demonstrated that heat treatment could reduce the osteoinductivity of DCB, and that H2O2 treatment was more effective than heat treatment.
Collapse
Affiliation(s)
- Shukun He
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Orthopedics, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ruonan Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuan Yao
- Department of Clinical Hematology, Faculty of Laboratory Medicine, Army Medical University, Chongqing, 400038, China
| | - Jing Cui
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huimin Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Zhu
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liangju Ning
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
20
|
Komori T. Regulation of Skeletal Development and Maintenance by Runx2 and Sp7. Int J Mol Sci 2024; 25:10102. [PMID: 39337587 PMCID: PMC11432631 DOI: 10.3390/ijms251810102] [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: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Runx2 (runt related transcription factor 2) and Sp7 (Sp7 transcription factor 7) are crucial transcription factors for bone development. The cotranscription factor Cbfb (core binding factor beta), which enhances the DNA-binding capacity of Runx2 and stabilizes the Runx2 protein, is necessary for bone development. Runx2 is essential for chondrocyte maturation, and Sp7 is partly involved. Runx2 induces the commitment of multipotent mesenchymal cells to osteoblast lineage cells and enhances the proliferation of osteoprogenitors. Reciprocal regulation between Runx2 and the Hedgehog, fibroblast growth factor (Fgf), Wnt, and parathyroid hormone-like hormone (Pthlh) signaling pathways and Dlx5 (distal-less homeobox 5) plays an important role in these processes. The induction of Fgfr2 (Fgf receptor 2) and Fgfr3 expression by Runx2 is important for the proliferation of osteoblast lineage cells. Runx2 induces Sp7 expression, and Runx2+ osteoprogenitors become Runx2+Sp7+ preosteoblasts. Sp7 induces the differentiation of preosteoblasts into osteoblasts without enhancing their proliferation. In osteoblasts, Runx2 is required for bone formation by inducing the expression of major bone matrix protein genes, including Col1a1 (collagen type I alpha 1), Col1a2, Spp1 (secreted phosphoprotein 1), Ibsp (integrin binding sialoprotein), and Bglap (bone gamma carboxyglutamate protein)/Bglap2. Bglap/Bglap2 (osteocalcin) regulates the alignment of apatite crystals parallel to collagen fibrils but does not function as a hormone that regulates glucose metabolism, testosterone synthesis, and muscle mass. Sp7 is also involved in Co1a1 expression and regulates osteoblast/osteocyte process formation, which is necessary for the survival of osteocytes and the prevention of cortical porosity. SP7 mutations cause osteogenesis imperfecta in rare cases. Runx2 is an important pathogenic factor, while Runx1, Runx3, and Cbfb are protective factors in osteoarthritis development.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Molecular Tumor Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
21
|
Yu XF, Teng B, Li JF, Zhang JV, Su Z, Ren PG. Novel Function of Osteocalcin in Chondrocyte Differentiation and Endochondral Ossification Revealed on a CRISPR/Cas9 bglap-bglap2 Deficiency Mouse Model. Int J Mol Sci 2024; 25:9945. [PMID: 39337434 PMCID: PMC11431882 DOI: 10.3390/ijms25189945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Endochondral ossification is the process by which cartilage is mineralized into bone, and is essential for the development of long bones. Osteocalcin (OCN), a protein abundant in bone matrix, also exhibits high expression in chondrocytes, especially hypertrophic chondrocytes, while its role in endochondral ossification remains unclear. Utilizing a new CRISPR/Cas9-mediated bglap-bglap2 deficiency (OCNem) mouse model generated in our laboratory, we provide the first evidence of OCN's regulatory function in chondrocyte differentiation and endochondral ossification. The OCNem mice exhibited significant delays in primary and secondary ossification centers compared to wild-type mice, along with increased cartilage length in growth plates and hypertrophic zones during neonatal and adolescent stages. These anomalies indicated that OCN deficiency disturbed endochondral ossification during embryonic and postnatal periods. Mechanism wise, OCN deficiency was found to increase chondrocyte differentiation and postpone vascularization process. Furthermore, bone marrow mesenchymal stromal cells (BMSCs) from OCNem mice demonstrated an increased capacity for chondrogenic differentiation. Transcriptional network analysis implicated that BMP and TGF-β signaling pathways were highly affected in OCNem BMSCs, which is closely associated with cartilage development and maintenance. This elucidation of OCN's function in chondrocyte differentiation and endochondral ossification contributes to a more comprehensive understanding of its impact on skeletal development and homeostasis.
Collapse
Affiliation(s)
- Xiang-Fang Yu
- Department of Endocrinology, Shenzhen Children’s Hospital, Shenzhen 518026, China;
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.T.); (J.-F.L.); (J.V.Z.)
| | - Bin Teng
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.T.); (J.-F.L.); (J.V.Z.)
| | - Jun-Feng Li
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.T.); (J.-F.L.); (J.V.Z.)
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jian V. Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (B.T.); (J.-F.L.); (J.V.Z.)
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhe Su
- Department of Endocrinology, Shenzhen Children’s Hospital, Shenzhen 518026, China;
| | - Pei-Gen Ren
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
- Center for Cancer Immunology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
22
|
Costa do Bomfim FR, Gonzalez Sella VR, Thomasini RL, Plapler H. Photobiomodulation Modulates Proliferation and Gene Expression Related to Calcium Signaling in Human Osteoblast Cells. J Lasers Med Sci 2024; 15:e45. [PMID: 39381787 PMCID: PMC11459251 DOI: 10.34172/jlms.2024.45] [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: 04/16/2024] [Accepted: 07/29/2024] [Indexed: 10/10/2024]
Abstract
Introduction: Photobiomodulation with low-level laser treatment can enhance bone formation by stimulating the cell division of osteoblasts and increasing the amount of protein deposition, thus encouraging the formation of new bone. The aim of this study was to evaluate the effects of photobiomodulation with a low-level laser on proliferation and gene expression related to calcium signaling in human osteoblasts. Methods: Osteoblastic cell lines of the hFOB1.19 lineage, human osteoblasts, were grown and assigned into two groups, control (C; n=78 cultured wells) and photobiomodulation (L; n=78 cultured wells) with n=6 per day of the experimental period. Cells were cultured (immature at 34 ºC), and after maturation at 37 ºC, group L cells were exposed to laser irradiation with a low-level laser device (gallium and aluminum arsenide), at a wavelength of 808 nm, a power output of 200 mW, and a power density of 200 mW/cm2. The energy delivered to the cells was 37 J/cm2, with a beam area of 0.02 mm2 and an exposure time of 5 seconds. This treatment was applied daily for a period of 13 days. Following this, the number of cells was counted, and RNA was isolated, measured, and then converted into cDNA for further quantification using a comparative Ct method with real-time polymerase chain reaction. The results were then subjected to statistical analysis through a Mann-Whitney test, with a significance level of P<0.05. Results: The cell count in the L group (37.25x10±4±22.02) was statistically higher compared to the control group (22.75x10±4±7.660) with a P value of 0.0259. The values of 2-ΔΔCt for S100A6, plasma membrane calcium ATPase (PMCA), and calmodulin genes indicated hyper-expression on the thirteenth day, while the osteocalcin gene showed hypo-expression. Conclusion: The study suggests that the photobiomodulation mechanism with a low-level laser may regulate gene expression in human osteoblasts in a dose-dependent and cumulative manner.
Collapse
Affiliation(s)
- Fernando Russo Costa do Bomfim
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
- Laboratory of Molecular Biology, Centro Universitário da Fundação Hermínio Ometto - FHO, Araras, SP, Brazil
| | - Valéria Regina Gonzalez Sella
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| | - Ronaldo Luis Thomasini
- Medicine Faculty, Universidade Federal dos Vales do Jequitinhonha e Mucuri - UFVJM, Diamantina, MG, Brazil
| | - Hélio Plapler
- Postgraduate Program in Interdisciplinary Surgical Science, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, SP, Brazil
| |
Collapse
|
23
|
McGarry S, Kover K, De Luca F. Thioredoxin Interacting Protein Expressed in Osteoblasts Mediates the Anti-Proliferative Effects of High Glucose and Modulates the Expression of Osteocalcin. J Bone Metab 2024; 31:209-218. [PMID: 39307521 PMCID: PMC11416880 DOI: 10.11005/jbm.2024.31.3.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Hyperglycemia is associated with impaired bone health in patients with diabetes mellitus. Although a direct detrimental effect of hyperglycemia on the bone has been previously reported, the specific molecular mediator(s) responsible for the inhibitory effect of high glucose levels on the bone remains unclear. We hypothesized that thioredoxin-interacting protein (Txnip), an essential mediator of oxidative stress, is such a mediator. METHODS We cultured MG-63 cells (immortalized human osteoblasts) with normal or high glucose concentrations and transfected them with scrambled or Txnip-specific small interfering RNA (siRNA). RESULTS High glucose levels increased Txnip expression and reduced MG-63 cell proliferation. The high-glucose level mediated reduction in cell proliferation was prevented in Txnip siRNA-transfected cells. In addition, we demonstrated that silencing Txnip mRNA expression in osteoblasts reduced the expression of the osteocalcin gene. Our results suggest that high glucose levels or silencing of Txnip mRNA expression may induce apoptosis in osteoblasts. CONCLUSIONS Our findings indicate that Txnip is an intracellular mediator of the anti-proliferative effects of extracellular high glucose levels on osteoblasts.
Collapse
Affiliation(s)
- Sarah McGarry
- Division of Endocrinology, Children’s Mercy Hospitals, Kansas City, MO,
USA
- Department of Pediatrics, University of Missouri-Kansas City-School of Medicine, Kansas City, MO,
USA
| | - Karen Kover
- Division of Endocrinology, Children’s Mercy Hospitals, Kansas City, MO,
USA
- Department of Pediatrics, University of Missouri-Kansas City-School of Medicine, Kansas City, MO,
USA
| | - Francesco De Luca
- Division of Endocrinology, Children’s Mercy Hospitals, Kansas City, MO,
USA
- Department of Pediatrics, University of Missouri-Kansas City-School of Medicine, Kansas City, MO,
USA
| |
Collapse
|
24
|
Wang S, Lei H, Mi Y, Ma P, Fan D. Chitosan and hyaluronic acid based injectable dual network hydrogels - Mediating antimicrobial and inflammatory modulation to promote healing of infected bone defects. Int J Biol Macromol 2024; 274:133124. [PMID: 38897505 DOI: 10.1016/j.ijbiomac.2024.133124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
In bone defects, infections lead to excessive inflammation, increased bacterial, and bone lysis, resulting in irregular wounds that hinder new bone regeneration. Injectable bioactive materials with adequate antimicrobial activity and strong osteogenic potential are urgently required to remedy irregular defects, eradicate bacteria, and facilitate the generation of new bone tissue. In this research, injectable dual-network composite hydrogels consisting of sulfated chitosan, oxidized hyaluronic acid, β-sodium glycerophosphate, and CuSr doped mesoporous bioactive glass loaded with bone morphogenetic protein (CuSrMBGBMP-2) were utilized for the first time to treat infectious bone defects. Initially, the hydrogel was injected into the wound at 37 °C with minimal invasion to establish a stable state and prevent hydrogel loss. Subsequently, sulfated chitosan eliminated bacteria at the wound site and facilitated cell proliferation with oxidized hyaluronic acid. Additionally, CuSrMBGBMP-2 strengthened antibacterial properties, regulated inflammatory reactions, promoted angiogenesis and osteogenic differentiation, addressing the deficiency in late-stage osteogenesis. Specifically, the injectable dual-network hydrogel based on chitosan and hyaluronic acid is minimally invasive, offering antibacterial, anti-inflammatory, pro-angiogenic, and bone regeneration properties. Therefore, this hydrogel with injectable dual network properties holds great promise for the treatment of bone infections in the future.
Collapse
Affiliation(s)
- Shang Wang
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Huan Lei
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Yu Mi
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Pei Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| |
Collapse
|
25
|
Szeliga A, Grymowicz M, Kostrzak A, Smolarczyk R, Bala G, Smolarczyk K, Meczekalski B, Suchta K. Bone: A Neglected Endocrine Organ? J Clin Med 2024; 13:3889. [PMID: 38999458 PMCID: PMC11242793 DOI: 10.3390/jcm13133889] [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: 05/25/2024] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/14/2024] Open
Abstract
Bone has traditionally been viewed in the context of its structural contribution to the human body. Foremost providing necessary support for mobility, its roles in supporting calcium homeostasis and blood cell production are often afterthoughts. Recent research has further shed light on the ever-multifaceted role of bone and its importance not only for structure, but also as a complex endocrine organ producing hormones responsible for the autoregulation of bone metabolism. Osteocalcin is one of the most important substances produced in bone tissue. Osteocalcin in circulation increases insulin secretion and sensitivity, lowers blood glucose, and decreases visceral adipose tissue. In males, it has also been shown to enhance testosterone production by the testes. Neuropeptide Y is produced by various cell types including osteocytes and osteoblasts, and there is evidence suggesting that peripheral NPY is important for regulation of bone formation. Hormonal disorders are often associated with abnormal levels of bone turnover markers. These include commonly used bone formation markers (bone alkaline phosphatase, osteocalcin, and procollagen I N-propeptide) and commonly used resorption markers (serum C-telopeptides of type I collagen, urinary N-telopeptides of type I collagen, and tartrate-resistant acid phosphatase type 5b). Bone, however, is not exclusively comprised of osseous tissue. Bone marrow adipose tissue, an endocrine organ often compared to visceral adipose tissue, is found between trabecula in the bone cortex. It secretes a diverse range of hormones, lipid species, cytokines, and other factors to exert diverse local and systemic effects.
Collapse
Affiliation(s)
- Anna Szeliga
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Monika Grymowicz
- Department of Gynecological Endocrinology, Warsaw Medical University, 00-315 Warsaw, Poland
| | - Anna Kostrzak
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Roman Smolarczyk
- Department of Gynecological Endocrinology, Warsaw Medical University, 00-315 Warsaw, Poland
| | - Gregory Bala
- UCD School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | | | - Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Katarzyna Suchta
- Department of Gynecological Endocrinology, Warsaw Medical University, 00-315 Warsaw, Poland
| |
Collapse
|
26
|
Li D, Liu C, Wang H, Li Y, Wang Y, An S, Sun S. The Role of Neuromodulation and Potential Mechanism in Regulating Heterotopic Ossification. Neurochem Res 2024; 49:1628-1642. [PMID: 38416374 DOI: 10.1007/s11064-024-04118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
Heterotopic ossification (HO) is a pathological process characterized by the aberrant formation of bone in muscles and soft tissues. It is commonly triggered by traumatic brain injury, spinal cord injury, and burns. Despite a wide range of evidence underscoring the significance of neurogenic signals in proper bone remodeling, a clear understanding of HO induced by nerve injury remains rudimentary. Recent studies suggest that injury to the nervous system can activate various signaling pathways, such as TGF-β, leading to neurogenic HO through the release of neurotrophins. These pathophysiological changes lay a robust groundwork for the prevention and treatment of HO. In this review, we collected evidence to elucidate the mechanisms underlying the pathogenesis of HO related to nerve injury, aiming to enhance our understanding of how neurological repair processes can culminate in HO.
Collapse
Affiliation(s)
- Dengju Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong First Medical University, Jinan, Shandong, China
| | - Changxing Liu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Haojue Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yunfeng Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yaqi Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Senbo An
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
| | - Shui Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
27
|
Lacombe J, Ferron M. Vitamin K-dependent carboxylation in β-cells and diabetes. Trends Endocrinol Metab 2024; 35:661-673. [PMID: 38429160 DOI: 10.1016/j.tem.2024.02.006] [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: 12/14/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024]
Abstract
Vitamin K is an essential micronutrient and a cofactor for the enzyme γ-glutamyl carboxylase, which adds a carboxyl group to specific glutamic acid residues in proteins transiting through the secretory pathway. Higher vitamin K intake has been linked to a reduced incidence of type 2 diabetes (T2D) in humans. Preclinical work suggests that this effect depends on the γ-carboxylation of specific proteins in β-cells, including endoplasmic reticulum Gla protein (ERGP), implicated in the control of intracellular Ca2+ levels. In this review we discuss these recent advances linking vitamin K and glucose metabolism, and argue that identification of γ-carboxylated proteins in β-cells is pivotal to better understand how vitamin K protects from T2D and to design targeted therapies for this disease.
Collapse
Affiliation(s)
- Julie Lacombe
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC, H2W 1R7, Canada.
| | - Mathieu Ferron
- Molecular Physiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC, H2W 1R7, Canada; Programme de Biologie Moléculaire, Université de Montréal, Montréal, QC, H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
28
|
Abudukelimu K, Aierken A, Tuerxuntayi A, Yilihamu Y, Abulizi S, Wufuer D, Dong H. Preliminary study on the preparation of antler powder/chitosan/β-glycerophosphate sodium/polyvinyl alcohol porous hydrogel scaffolds and their osteogenic effects. Front Bioeng Biotechnol 2024; 12:1421718. [PMID: 38988866 PMCID: PMC11233688 DOI: 10.3389/fbioe.2024.1421718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction: The production of bone-like structural scaffolds through bone tissue engineering technology is a promising method for bone regeneration to repair bone defects. Deer antler, an easily harvested and abundantly sourced initial bone tissue structure, resembles the composition and structure of human cancellous bone and can serve as a new material for allogeneic bone transplantation. Methods: This study involved the preparation and characterization of antler powder/chitosan/β-glycerophosphate sodium/polyvinyl alcohol (AP/CS/β-GP/PVA) porous hydrogel scaffolds to verify their material properties and osteogenic mechanisms. The microstructure, hydrophilicity, and mechanical properties of the scaffolds were studied using Scanning Electron Microscopy (SEM), contact angle measurement, and a universal material testing machine. The interactions between the various components were investigated using Fourier-Transform Infrared Spectroscopy (FTIR). Biocompatibility, osteogenic properties, and expression of osteogenesis-related proteins of the scaffolds were evaluated through Cell Counting Kit-8 (CCK-8) assays, alkaline phosphatase staining, Alizarin Red staining, live/dead cell staining, and Western blot analysis. Results: The results showed that as the content of deer antler powder increased, both the hydrophilicity and mechanical properties of the scaffold materials improved, while the porosity slightly decreased with an increase in deer antler powder content. Cell culture experiments demonstrated that scaffolds with a higher proportion of deer antler powder were beneficial for the proliferation and differentiation of mouse pre-osteoblast (MC3T3-E1) cells, with the scaffolds containing 10% and 8% deer antler powder showing the best effects. The upregulation of RUNX2, OCN, OSX, and OPN protein expression may promote differentiation. Discussion: Therefore, the AP/CS/β-GP/PVA hydrogel scaffolds have the potential to become a promising biomaterial for bone tissue engineering.
Collapse
Affiliation(s)
- Kudelaiti Abudukelimu
- Department of Prosthodontics, The First Affiliated Hospital (Affiliated Stomatological Hospital) of Xinjiang Medical University, Urumqi, China
- People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Aikepaer Aierken
- People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | | | - Yilizhati Yilihamu
- College of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Saierdaer Abulizi
- People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Duolikun Wufuer
- People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Hongbin Dong
- Department of Prosthodontics, The First Affiliated Hospital (Affiliated Stomatological Hospital) of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
29
|
Ravazzano L, Colaianni G, Tarakanova A, Xiao YB, Grano M, Libonati F. Multiscale and multidisciplinary analysis of aging processes in bone. NPJ AGING 2024; 10:28. [PMID: 38879533 PMCID: PMC11180112 DOI: 10.1038/s41514-024-00156-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/07/2024] [Indexed: 06/19/2024]
Abstract
The world population is increasingly aging, deeply affecting our society by challenging our healthcare systems and presenting an economic burden, thus turning the spotlight on aging-related diseases: exempli gratia, osteoporosis, a silent disease until you suddenly break a bone. The increase in bone fracture risk with age is generally associated with a loss of bone mass and an alteration in the skeletal architecture. However, such changes cannot fully explain increased fragility with age. To successfully tackle age-related bone diseases, it is paramount to comprehensively understand the fundamental mechanisms responsible for tissue degeneration. Aging mechanisms persist at multiple length scales within the complex hierarchical bone structure, raising the need for a multiscale and multidisciplinary approach to resolve them. This paper aims to provide an overarching analysis of aging processes in bone and to review the most prominent outcomes of bone aging. A systematic description of different length scales, highlighting the corresponding techniques adopted at each scale and motivating the need for combining diverse techniques, is provided to get a comprehensive description of the multi-physics phenomena involved.
Collapse
Affiliation(s)
- Linda Ravazzano
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Via Rubattino 81, Milano, 20134, Italy
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11, Bari, 70124, Italy
| | - Anna Tarakanova
- School of Mechanical, Aerospace, and Manufacturing Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, 06269, CT, USA
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, CT, 06269, Storrs, USA
| | - Yu-Bai Xiao
- School of Mechanical, Aerospace, and Manufacturing Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, 06269, CT, USA
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11, Bari, 70124, Italy
| | - Flavia Libonati
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Via Rubattino 81, Milano, 20134, Italy.
- Department of Mechanical, Energy, Management and Transport Engineering - DIME, University of Genova, Via all'Opera Pia 15, Genova, 16145, Italy.
| |
Collapse
|
30
|
Wolfe PN, Stoker AM, Leary E, Crist BD, Bozynski CC, Cook JL. Evaluation of Serum and Urine Biomarker Panels for Developmental Dysplasia of the Hip Prior to Onset of Secondary Osteoarthritis. Cartilage 2024; 15:164-174. [PMID: 37051936 PMCID: PMC11368892 DOI: 10.1177/19476035231163032] [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: 01/13/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 04/14/2023] Open
Abstract
OBJECTIVE Evaluate serum and urine biomarker panels for their capabilities in discriminating between individuals (13- to 34-years-olds) with healthy hips versus those with developmental dysplasia of the hip (DDH) prior to diagnosis of secondary hip osteoarthritis (OA). DESIGN Urine and serum were collected from individuals (15-33 years old) with DDH, prior to and following diagnosis of hip OA, and from age-matched healthy-hip controls. Samples were analyzed for panels of protein biomarkers with potential for differentiation of hip status using receiver operator characteristic curve (area under curve [AUC]) assessments. RESULTS Multiple urine and serum biomarker panels effectively differentiated individuals with DDH from healthy-hip controls in a population at risk for developing secondary hip OA with the best performing panel demonstrating an AUC of 0.959. The panel comprised of two serum and two urinary biomarkers provided the highest combined values for sensitivity, 0.85, and specificity, 1.00, while a panel of four serum biomarkers provided the highest sensitivity, 0.93, while maintaining adequate specificity, 0.71. CONCLUSION Results of this study indicate that panels of protein biomarkers measured in urine and serum may be able to differentiate young adults with DDH from young adults with healthy hips. These data suggest the potential for clinical application of a routine diagnostic method for cost-effective and timely screening for DDH in at-risk populations. Further development and validation of these biomarker panels may result in highly sensitive and specific tools for early diagnosis, staging, and prognostication of DDH, as well as treatment decision making and monitoring capabilities. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
- Preston N. Wolfe
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - Aaron M. Stoker
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - Emily Leary
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
- School of Medicine, University of Missouri, Columbia, MO, USA
| | - Brett D. Crist
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| | - Chantelle C. Bozynski
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - James L. Cook
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| |
Collapse
|
31
|
Nowicki JK, Jakubowska-Pietkiewicz E. Osteocalcin: Beyond Bones. Endocrinol Metab (Seoul) 2024; 39:399-406. [PMID: 38803289 PMCID: PMC11220208 DOI: 10.3803/enm.2023.1895] [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: 11/28/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 05/29/2024] Open
Abstract
Apart from basic roles such as supporting the body, protecting internal organs, and storing calcium, the skeletal system also performs hormonal functions. In recent years, several reports have been published on proteins secreted by bones and their impact on the homeostasis of the entire body. These proteins include fibroblast growth factor 23, sclerostin, lipocalin 2, and osteocalcin. Osteocalcin, the most abundant non-collagenous protein in bone tissue, is routinely measured as a clinical marker for diagnosing bone metabolism disorders. Its molecule undergoes numerous transformations, with decarboxylation being the critical process. Decarboxylation occurs in the acidic environment typical of bone resorption, facilitating the release of the molecule into the bloodstream and enabling its hormonal action. Decarboxylated osteocalcin promotes insulin secretion and stimulates the proliferation of pancreatic islet β-cells. It also plays a role in reducing the accumulation of visceral fat and decreasing fat storage in the liver. Furthermore, decarboxylated osteocalcin levels are inversely correlated with fasting serum glucose levels, total body fat, visceral fat area, and body mass index. Apart from its role in energy metabolism, osteocalcin affects testosterone production and the synthesis of glucagon-like peptide-1. It is also actively involved in muscle-bone crosstalk and influences cognitive function.
Collapse
Affiliation(s)
- Jakub Krzysztof Nowicki
- Department of Pediatrics, Neonatal Pathology and Metabolic Bone Diseases, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
32
|
Bharath Kumar BS, Mallick S, Manjunathachar HV, Shashank CG, Sharma A, Nagoorvali D, Soren S, Jadhav VG, Pandita S. In vitro effects of uncarboxylated osteocalcin on buffalo Leydig cell steroidogenesis. Vet Res Commun 2024; 48:1423-1433. [PMID: 38305958 DOI: 10.1007/s11259-024-10320-4] [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: 11/21/2023] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Uncarboxylated osteocalcin (UcOCN), a bone derived circulating protein, has been demonstrated to influence steroidogenesis in testicular Leydig cells of murine and human species. However, the role of UcOCN in testosterone biosynthesis remains unexplored in domestic animals. The present study aimed to investigate the impact of UcOCN on the expressions of steroidogenic genes (HSD3β1, HSD3β6, CYP17A1, CYP11A1), testosterone production and GPRC6A receptor localization in buffalo Leydig cells. Leydig cells from the testes of adult Murrah buffalo were isolated, with an average cell count and viability after digestion and Percoll enrichment of 1.43 × 106 cells/g of testes and 78.5%, respectively. Immunophenotyping of Percoll-enriched cell suspension by flow cytometry showed populations of Leydig cells ranging between 69 and 73.9%. Immunostaining confirmed the presence of GPRC6A receptors and CYP11A1 positive Leydig cells. When these cells were cultured and incubated with varying levels of UcOCN (6, 12, 24, and 48 ng/ml) and LH, there was a significant (P < 0.01) increase in testosterone production and up-regulation (P < 0.05) of CYP11A1, CYP17A1, HSD3β1 and HSD3β6 gene expression. In summary, the present study underscored the effects of UcOCN on testosterone biosynthesis, expression of crucial steroidogenic genes and interaction with GPRC6A receptors in buffalo Leydig cells, emphasizing its potential implications in andrology.
Collapse
Affiliation(s)
- B S Bharath Kumar
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
- Department of Animal Husbandry, Government of Karnataka, Bengaluru, Karnataka, India.
| | - Smrutirekha Mallick
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
- ICAR-National Institute on Foot and Mouth Disease, Bhubaneswar, Odisha, 752050, India.
| | - H V Manjunathachar
- ICAR-Indian Veterinary Research Institute, Bareilly, U.P., 243122, India
- ICMR-National Institute of Traditional Medicine, Belagavi, Karnataka, 590010, India
| | - C G Shashank
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Ankur Sharma
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | | | - Simson Soren
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | | | - Sujata Pandita
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
33
|
Guo X, Yang YY, Zhou R, Tian G, Shan C, Liu JM, Li R. Causal effect of blood osteocalcin on the risk of Alzheimer's disease and the mediating role of energy metabolism. Transl Psychiatry 2024; 14:205. [PMID: 38769320 PMCID: PMC11106250 DOI: 10.1038/s41398-024-02924-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
Growing evidence suggests an association between osteocalcin (OCN), a peptide derived from bone and involved in regulating glucose and lipid metabolism, and the risk of Alzheimer's disease (AD). However, the causality of these associations and the underlying mechanisms remain uncertain. We utilized a Mendelian randomization (MR) approach to investigate the causal effects of blood OCN levels on AD and to assess the potential involvement of glucose and lipid metabolism. Independent instrumental variables strongly associated (P < 5E-08) with blood OCN levels were obtained from three independent genome-wide association studies (GWAS) on the human blood proteome (N = 3301 to 35,892). Two distinct summary statistics datasets on AD from the International Genomics of Alzheimer's Project (IGAP, N = 63,926) and a recent study including familial-proxy AD patients (FPAD, N = 472,868) were used. Summary-level data for fasting glucose (FG), 2h-glucose post-challenge, fasting insulin, HbA1c, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, total cholesterol (TC), and triglycerides were incorporated to evaluate the potential role of glucose and lipid metabolism in mediating the impact of OCN on AD risk. Our findings consistently demonstrate a significantly negative correlation between genetically determined blood OCN levels and the risk of AD (IGAP: odds ratio [OR, 95%CI] = 0.83[0.72-0.96], P = 0.013; FPAD: OR = 0.81 [0.70-0.93], P = 0.002). Similar estimates with the same trend direction were obtained using other statistical approaches. Furthermore, employing multivariable MR analysis, we found that the causal relationship between OCN levels and AD was disappeared after adjustment of FG and TC (IGAP: OR = 0.97[0.80-1.17], P = 0.753; FPAD: OR = 0.98 [0.84-1.15], P = 0.831). There were no apparent instances of horizontal pleiotropy, and leave-one-out analysis showed good stability of the estimates. Our study provides evidence supporting a protective effect of blood OCN levels on AD, which is primarily mediated through regulating FG and TC levels. Further studies are warranted to elucidate the underlying physio-pathological mechanisms.
Collapse
Affiliation(s)
- Xingzhi Guo
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
- Department of Geriatric Neurology, the Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Yu-Ying Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Rong Zhou
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
- Department of Geriatric Neurology, the Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Ge Tian
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Chang Shan
- Department of Endocrinology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Rui Li
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China.
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China.
- Department of Geriatric Neurology, the Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
34
|
Arakil N, Akhund SA, Elaasser B, Mohammad KS. Intersecting Paths: Unraveling the Complex Journey of Cancer to Bone Metastasis. Biomedicines 2024; 12:1075. [PMID: 38791037 PMCID: PMC11117796 DOI: 10.3390/biomedicines12051075] [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: 03/17/2024] [Revised: 04/27/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
The phenomenon of bone metastases presents a significant challenge within the context of advanced cancer treatments, particularly pertaining to breast, prostate, and lung cancers. These metastatic occurrences stem from the dissemination of cancerous cells into the bone, thereby interrupting the equilibrium between osteoblasts and osteoclasts. Such disruption results in skeletal complications, adversely affecting patient morbidity and quality of life. This review discusses the intricate interplay between cancer cells and the bone microenvironment, positing the bone not merely as a passive recipient of metastatic cells but as an active contributor to cancer progression through its distinctive biochemical and cellular makeup. A thorough examination of bone structure and the dynamics of bone remodeling is undertaken, elucidating how metastatic cancer cells exploit these processes. This review explores the genetic and molecular pathways that underpin the onset and development of bone metastases. Particular emphasis is placed on the roles of cytokines and growth factors in facilitating osteoclastogenesis and influencing osteoblast activity. Additionally, this paper offers a meticulous critique of current diagnostic methodologies, ranging from conventional radiography to advanced molecular imaging techniques, and discusses the implications of a nuanced understanding of bone metastasis biology for therapeutic intervention. This includes the development of targeted therapies and strategies for managing bone pain and other skeletal-related events. Moreover, this review underscores the imperative of ongoing research efforts aimed at identifying novel therapeutic targets and refining management approaches for bone metastases. It advocates for a multidisciplinary strategy that integrates advancements in medical oncology and radiology with insights derived from molecular biology and genetics, to enhance prognostic outcomes and the quality of life for patients afflicted by this debilitating condition. In summary, bone metastases constitute a complex issue that demands a comprehensive and informed approach to treatment. This article contributes to the ongoing discourse by consolidating existing knowledge and identifying avenues for future investigation, with the overarching objective of ameliorating patient care in the domain of oncology.
Collapse
Affiliation(s)
| | | | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia; (N.A.); (S.A.A.); (B.E.)
| |
Collapse
|
35
|
Paracha N, Mastrokostas P, Kello E, Gedailovich Y, Segall D, Rizzo A, Mitelberg L, Hassan N, Dowd TL. Osteocalcin improves glucose tolerance, insulin sensitivity and secretion in older male mice. Bone 2024; 182:117048. [PMID: 38378083 DOI: 10.1016/j.bone.2024.117048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/19/2024] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
Osteocalcin deficient mice (OC-/-), on a mixed 129/BL6J background, were reported to show glucose intolerance, insulin insensitivity and reduced insulin secretion at 1-6 mos of age. This is controversial as two studies in OC-/- mice on different backgrounds (C3H/BL6 (5-6 mos.) and C57BL/6N (5 and 9 mos.)) found no effect on glucose metabolism. To determine the role of OC in glucose metabolism we conducted glucose tolerance tests (GTT), insulin tolerances tests (ITT) and glucose stimulated insulin secretion (GSIS) on 6 and 9.5 month-old male OC-/- and OC+/+ mice on a pure C57BL/6J background and fed a normal chow diet. All results were analyzed with a two-way repeated measures ANOVA. The GTT results showed no effect on males at 6 months of age but glucose intolerance was significantly increased (p < 0.05) in male OC-/- mice at 9.5 months of age. The ITT results indicated significantly increased insulin resistance in male OC-/- mice. Glucose stimulated insulin secretion (GSIS) showed insulin significantly (p < 0.05) reduced in OC-/- at several time points. Mouse Osteocalcin injected into OC-/- mice decreased the glucose level. Our results confirm the role of OC in glucose metabolism and insulin sensitivity and demonstrate a role in insulin secretion in older male mice on a C57BL/6J background. Differences in background, age, or experimental procedures could explain controversial results. A delayed onset of the effect of OC on glucose metabolism at 9.5 months in male C57BL/6J mice highlights the importance of background on phenotype. Consideration of genetic background and age may be beneficial for human studies on osteocalcin and glucose homeostasis and may be relevant to the elderly where osteocalcin is reduced.
Collapse
Affiliation(s)
- Noorulain Paracha
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Paul Mastrokostas
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Evan Kello
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Yosef Gedailovich
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Devorah Segall
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Alexis Rizzo
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Lawrence Mitelberg
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Naif Hassan
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America
| | - Terry Lynne Dowd
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY 11210, United States of America; Ph.D. Program in Chemistry and Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America.
| |
Collapse
|
36
|
Elseweidy MM, Ali AEM, Hassanin SM, Mahmoud YK. Empagliflozin ameliorates liver fibrosis in NASH rat model via targeting hepatic NF-κB/SOX9/OPN signaling and osteocalcin level. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3449-3459. [PMID: 37962587 PMCID: PMC11074015 DOI: 10.1007/s00210-023-02826-6] [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: 09/07/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) may be associated with tissue fibrotic changes and can be treated via different therapeutic tools which may however either initiate weak or long-term side effects that minimize its use. Empagliflozin (EMPA) is an oral anti-diabetic drug which has characteristic effects during hepatic steatosis regarding lipid accumulation and insulin resistance. In this study, we aimed to investigate an additional mechanism through which EMPA can exert and potentiate its anti-inflammatory and anti-fibrotic effects in NASH rat model. Male Wistar albino rats fed on high fat diet (HFD) and 20% fructose in drinking water for 18 weeks and received EMPA (30 mg/kg/day, orally) starting from week 11. Body and liver weights, homeostatic model assessment of insulin resistance (HOMA-IR), lipid profile, liver function tests, other biochemical and histological parameters were determined. HFD joined with fructose intake significantly increased body and liver weights, HOMA-IR value, hepatic inflammatory and fibrotic markers, liver transaminases, hepatic expression of nuclear factor-kappa B (NF-κB), sex determining region Y box 9 (SOX 9), and osteopontin (OPN) with significant decrease in hepatic osteocalcin (OCN). Intense hepatic lesions with severe microsteatosis and deposition of collagen fibers were clearly observed. Effectively, EMPA restored the normal liver functions, downregulated hepatic inflammatory cytokines, NF-κB, SOX 9, OPN, and increased OCN level. These results highlight another pathway illustrated the anti-fibrotic effects of EMPA against liver fibrosis probably through downregulation of NF-κB/SOX 9/OPN signaling along with upregulation of hepatic OCN which may potentiate the valuable anti-inflammatory and anti-fibrotic effects of EMPA.
Collapse
Affiliation(s)
- Mohamed M Elseweidy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Abd El-Monem Ali
- Pathology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Sara M Hassanin
- Zagazig University Hospitals, Zagazig University, Zagazig, Egypt
| | - Yasmin K Mahmoud
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
37
|
Wirsig K, Bacova J, Richter RF, Hintze V, Bernhardt A. Cellular response of advanced triple cultures of human osteocytes, osteoblasts and osteoclasts to high sulfated hyaluronan (sHA3). Mater Today Bio 2024; 25:101006. [PMID: 38445011 PMCID: PMC10912908 DOI: 10.1016/j.mtbio.2024.101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Bone remodelling, important for homeostasis and regeneration involves the controlled action of osteoblasts, osteocytes and osteoclasts. The present study established a three-dimensional human in vitro bone model as triple culture with simultaneously differentiating osteocytes and osteoclasts, in the presence of osteoblasts. Since high sulfated hyaluronan (sHA3) was reported as a biomaterial to enhance osteogenesis as well as to dampen osteoclastogenesis, the triple culture was exposed to sHA3 to investigate cellular responses compared to the respective bone cell monocultures. Osteoclast formation and marker expression was stimulated by sHA3 only in triple culture. Osteoprotegerin (OPG) gene expression and protein secretion, but not receptor activator of NF-κB ligand (RANKL) or sclerostin (SOST), were strongly enhanced, suggesting an important role of sHA3 itself in osteoclastogenesis with other targets than indirect modulation of the RANKL/OPG ratio. Furthermore, sHA3 upregulated osteocalcin (BGLAP) in osteocytes and osteoblasts in triple culture, while alkaline phosphatase (ALP) was downregulated.
Collapse
Affiliation(s)
- Katharina Wirsig
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
| | - Jana Bacova
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 53210 Pardubice, Czech Republic
| | - Richard F. Richter
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
| | - Vera Hintze
- Max Bergmann Center of Biomaterials, Institute of Material Science, TUD University of Technology, Budapester Str. 27, 01069, Dresden, Germany
| | - Anne Bernhardt
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
| |
Collapse
|
38
|
Martiniakova M, Biro R, Kovacova V, Babikova M, Zemanova N, Mondockova V, Omelka R. Current knowledge of bone-derived factor osteocalcin: its role in the management and treatment of diabetes mellitus, osteoporosis, osteopetrosis and inflammatory joint diseases. J Mol Med (Berl) 2024; 102:435-452. [PMID: 38363329 PMCID: PMC10963459 DOI: 10.1007/s00109-024-02418-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 02/17/2024]
Abstract
Osteocalcin (OC) is the most abundant non-collagenous and osteoblast-secreted protein in bone. It consists of two forms such as carboxylated OC (cOC) and undercarboxylated OC (ucOC). While cOC promotes bone mineralization and increases bone strength, ucOC is regarded an endocrinologically active form that may have several functions in multiple end organs and tissues. Total OC (tOC) includes both of these forms (cOC and ucOC) and is considered a marker of bone turnover in clinical settings. Most of the data on OC is limited to preclinical studies and therefore may not accurately reflect the situation in clinical conditions. For the stated reason, the aim of this review was not only to summarize current knowledge of all forms of OC and characterize its role in diabetes mellitus, osteoporosis, osteopetrosis, inflammatory joint diseases, but also to provide new interpretations of its involvement in the management and treatment of aforementioned diseases. In this context, special emphasis was placed on available clinical trials. Significantly lower levels of tOC and ucOC could be associated with the risk of type 2 diabetes mellitus. On the contrary, tOC level does not seem to be a good indicator of high bone turnover status in postmenopausal osteoporosis, osteoarthritis and rheumatoid arthritis. The associations between several pharmacological drugs used to treat all disorders mentioned above and OC levels have also been provided. From this perspective, OC may serve as a medium through which certain medications can influence glucose metabolism, body weight, adiponectin secretion, and synovial inflammation.
Collapse
Affiliation(s)
- Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Roman Biro
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Martina Babikova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Nina Zemanova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, Tr. A. Hlinku 1, 949 01, Nitra, Slovakia.
| |
Collapse
|
39
|
Cheng M, Nie Y, Song M, Chen F, Yu Y. Forkhead box O proteins: steering the course of stem cell fate. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:7. [PMID: 38466341 DOI: 10.1186/s13619-024-00190-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
Stem cells are pivotal players in the intricate dance of embryonic development, tissue maintenance, and regeneration. Their behavior is delicately balanced between maintaining their pluripotency and differentiating as needed. Disruptions in this balance can lead to a spectrum of diseases, underscoring the importance of unraveling the complex molecular mechanisms that govern stem cell fate. Forkhead box O (FOXO) proteins, a family of transcription factors, are at the heart of this intricate regulation, influencing a myriad of cellular processes such as survival, metabolism, and DNA repair. Their multifaceted role in steering the destiny of stem cells is evident, as they wield influence over self-renewal, quiescence, and lineage-specific differentiation in both embryonic and adult stem cells. This review delves into the structural and regulatory intricacies of FOXO transcription factors, shedding light on their pivotal roles in shaping the fate of stem cells. By providing insights into the specific functions of FOXO in determining stem cell fate, this review aims to pave the way for targeted interventions that could modulate stem cell behavior and potentially revolutionize the treatment and prevention of diseases.
Collapse
Affiliation(s)
- Mengdi Cheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Yujie Nie
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Min Song
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China.
- Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
40
|
Hino H, Kondo S, Kuroda J. In vivo imaging of bone collagen dynamics in zebrafish. Bone Rep 2024; 20:101748. [PMID: 38525199 PMCID: PMC10959726 DOI: 10.1016/j.bonr.2024.101748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/21/2024] [Accepted: 03/03/2024] [Indexed: 03/26/2024] Open
Abstract
Type I collagen plays a pivotal role in shaping bone morphology and determining its physical properties by serving as a template for ossification. Nevertheless, the mechanisms underlying bone collagen formation, particularly the principles governing its orientation, remain unknown owing to the lack of a method that enables continuous in vivo observations. To address this challenge, we constructed a method to visualize bone collagen by tagging with green fluorescent protein (GFP) in zebrafish and observed the interactions between osteoblasts and collagen fibers during bone formation in vivo. When collagen type I alpha 2 chain (Col1a2)-GFP was expressed under the control of the osteoblast-specific promoters osx or osc in zebrafish, bone collagen was observed clearly enough to identify its localization, whereas collagen from other organs was not. Therefore, we determined that this method was of sufficient quality for the detailed in vivo observation of bone collagen. Next, bone collagen in the scales, fin rays, and opercular bones of zebrafish was observed in detail, when bone formation is more active. High-magnification imaging showed that Col1a2-GFP can visualize collagen sufficiently to analyze the collagen fiber orientation and microstructure of bones. Furthermore, by simultaneously observation of bone collagen and osteoblasts, we successfully observed dynamic changes in the morphology and position of osteoblasts from the early stages of bone formation. It was also found that the localization pattern and orientation of bone collagen significantly differed depending on the choice of the expression promoter. Both promoters (osx and osc) used in this study are osteoblast-specific, but their Col1a2-GFP localizing regions within the bone were exclusive, with osx region localizing mainly to the outer edge of the bone and osc region localizing to the central area of the bone. This suggests the existence of distinct osteoblast subpopulations with different gene expression profiles, each of which may play a unique role in osteogenesis. These findings would contribute to a better understanding of the mechanisms governing bone collagen formation by osteoblasts.
Collapse
Affiliation(s)
- Hiromu Hino
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Shigeru Kondo
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Junpei Kuroda
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
41
|
Sidgwick GP, Weston R, Mahmoud AM, Schiro A, Serracino-Inglott F, Tandel SM, Skeoch S, Bruce IN, Jones AM, Alexander MY, Wilkinson FL. Novel Glycomimetics Protect against Glycated Low-Density Lipoprotein-Induced Vascular Calcification In Vitro via Attenuation of the RAGE/ERK/CREB Pathway. Cells 2024; 13:312. [PMID: 38391925 PMCID: PMC10887290 DOI: 10.3390/cells13040312] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Heparan sulphate (HS) can act as a co-receptor on the cell surface and alterations in this process underpin many pathological conditions. We have previously described the usefulness of mimics of HS (glycomimetics) in protection against β-glycerophosphate-induced vascular calcification and in the restoration of the functional capacity of diabetic endothelial colony-forming cells in vitro. This study aims to investigate whether our novel glycomimetic compounds can attenuate glycated low-density lipoprotein (g-LDL)-induced calcification by inhibiting RAGE signalling within the context of critical limb ischemia (CLI). We used an established osteogenic in vitro vascular smooth muscle cell (VSMC) model. Osteoprotegerin (OPG), sclerostin and glycation levels were all significantly increased in CLI serum compared to healthy controls, while the vascular calcification marker osteocalcin (OCN) was down-regulated in CLI patients vs. controls. Incubation with both CLI serum and g-LDL (10 µg/mL) significantly increased VSMC calcification vs. controls after 21 days, with CLI serum-induced calcification apparent after only 10 days. Glycomimetics (C2 and C3) significantly inhibited g-LDL and CLI serum-induced mineralisation, as shown by a reduction in alizarin red (AR) staining and alkaline phosphatase (ALP) activity. Furthermore, secretion of the osteogenic marker OCN was significantly reduced in VSMCs incubated with CLI serum in the presence of glycomimetics. Phosphorylation of cyclic AMP response element-binding protein (CREB) was significantly increased in g-LDL-treated cells vs. untreated controls, which was attenuated with glycomimetics. Blocking CREB activation with a pharmacological inhibitor 666-15 replicated the protective effects of glycomimetics, evidenced by elevated AR staining. In silico molecular docking simulations revealed the binding affinity of the glycomimetics C2 and C3 with the V domain of RAGE. In conclusion, these findings demonstrate that novel glycomimetics, C2 and C3 have potent anti-calcification properties in vitro, inhibiting both g-LDL and CLI serum-induced VSMC mineralisation via the inhibition of LDLR, RAGE, CREB and subsequent expression of the downstream osteogenic markers, ALP and OCN.
Collapse
Affiliation(s)
- Gary P. Sidgwick
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
| | - Ria Weston
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
| | - Ayman M. Mahmoud
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
| | - Andrew Schiro
- Cardiovascular Research Institute, University of Manchester, Manchester M13 9PL, UK;
- Vascular Unit, Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - Ferdinand Serracino-Inglott
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
- Cardiovascular Research Institute, University of Manchester, Manchester M13 9PL, UK;
- Vascular Unit, Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - Shikha M. Tandel
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
| | - Sarah Skeoch
- Centre for Epidemiology Versus Arthritis, University of Manchester, Manchester M13 9PL, UK; (S.S.); (I.N.B.)
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
- Royal National Hospital for Rheumatic Diseases, Bath BA1 1RL, UK
| | - Ian N. Bruce
- Centre for Epidemiology Versus Arthritis, University of Manchester, Manchester M13 9PL, UK; (S.S.); (I.N.B.)
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Alan M. Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
- School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK
| | - M. Yvonne Alexander
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
| | - Fiona L. Wilkinson
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK (R.W.); (A.M.M.); (F.S.-I.); (S.M.T.); (A.M.J.); (M.Y.A.)
| |
Collapse
|
42
|
Chen S, Croft AS, Bigdon S, Albers CE, Li Z, Gantenbein B. Conditioned Medium of Intervertebral Disc Cells Inhibits Osteo-Genesis on Autologous Bone-Marrow-Derived Mesenchymal Stromal Cells and Osteoblasts. Biomedicines 2024; 12:376. [PMID: 38397978 PMCID: PMC10886592 DOI: 10.3390/biomedicines12020376] [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: 12/13/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Low back pain (LBP) is associated with the degeneration of human intervertebral discs (IVDs). Despite progress in the treatment of LBP through spinal fusion, some cases still end in non-fusion after the removal of the affected IVD tissue. In this study, we investigated the hypothesis that the remaining IVD cells secrete BMP inhibitors that are sufficient to inhibit osteogenesis in autologous osteoblasts (OBs) and bone marrow mesenchymal stem cells (MSCs). A conditioned medium (CM) from primary human IVD cells in 3D alginate culture was co-cultured with seven donor-matched OB and MSCs. After ten days, osteogenesis was quantified at the transcript level using qPCR to measure the expression of bone-related genes and BMP antagonists, and at the protein level by alkaline phosphatase (ALP) activity. Additionally, cells were evaluated histologically using alizarin red (ALZR) staining on Day 21. For judging ALP activity and osteogenesis, the Noggin expression in samples was investigated to uncover the potential causes. The results after culture with the CM showed significantly decreased ALP activity and the inhibition of the calcium deposit formation in alizarin red staining. Interestingly, no significant changes were found among most bone-related genes and BMP antagonists in OBs and MSCs. Noteworthy, Noggin was relatively expressed higher in human IVD cells than in autologous OBs or MSCs (relative to autologous OB, the average fold change was in 6.9, 10.0, and 6.3 in AFC, CEPC, and NPC, respectively; and relative to autologous MSC, the average fold change was 2.3, 3.4, and 3.2, in AFC, CEPC, and NPC, respectively). The upregulation of Noggin in residual human IVDs could potentially inhibit the osteogenesis of autologous OB and MSC, thus inhibiting the postoperative spinal fusion after discectomy surgery.
Collapse
Affiliation(s)
- Shuimu Chen
- Tissue Engineering for Orthopedics & Mechanobiology (TOM), Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Andreas S Croft
- Tissue Engineering for Orthopedics & Mechanobiology (TOM), Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Sebastian Bigdon
- Department of Orthopedic Surgery & Traumatology, Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Christoph E Albers
- Department of Orthopedic Surgery & Traumatology, Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Zhen Li
- AO Research Institute Davos, 7270 Davos, Switzerland
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopedics & Mechanobiology (TOM), Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008 Bern, Switzerland
- Department of Orthopedic Surgery & Traumatology, Inselspital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
43
|
Correa Pinto Junior D, Canal Delgado I, Yang H, Clemenceau A, Corvelo A, Narzisi G, Musunuri R, Meyer Berger J, Hendricks LE, Tokumura K, Luo N, Li H, Oury F, Ducy P, Yadav VK, Li X, Karsenty G. Osteocalcin of maternal and embryonic origins synergize to establish homeostasis in offspring. EMBO Rep 2024; 25:593-615. [PMID: 38228788 PMCID: PMC10897216 DOI: 10.1038/s44319-023-00031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024] Open
Abstract
Many physiological osteocalcin-regulated functions are affected in adult offspring of mothers experiencing unhealthy pregnancy. Furthermore, osteocalcin signaling during gestation influences cognition and adrenal steroidogenesis in adult mice. Together these observations suggest that osteocalcin may broadly function during pregnancy to determine organismal homeostasis in adult mammals. To test this hypothesis, we analyzed in unchallenged wildtype and Osteocalcin-deficient, newborn and adult mice of various genotypes and origin maintained on different genetic backgrounds, the functions of osteocalcin in the pancreas, liver and testes and their molecular underpinnings. This analysis revealed that providing mothers are Osteocalcin-deficient, Osteocalcin haploinsufficiency in embryos hampers insulin secretion, liver gluconeogenesis, glucose homeostasis, testes steroidogenesis in adult offspring; inhibits cell proliferation in developing pancreatic islets and testes; and disrupts distinct programs of gene expression in these organs and in the brain. This study indicates that osteocalcin exerts dominant functions in most organs it influences. Furthermore, through their synergistic regulation of multiple physiological functions, osteocalcin of maternal and embryonic origins contributes to the establishment and maintenance of organismal homeostasis in newborn and adult offspring.
Collapse
Affiliation(s)
- Danilo Correa Pinto Junior
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Isabella Canal Delgado
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Haiyang Yang
- Guangdong Provincial Key Laboratory of Brain Connectome, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Alisson Clemenceau
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | | | | | - Julian Meyer Berger
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Lauren E Hendricks
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Na Luo
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Hongchao Li
- Guangdong Provincial Key Laboratory of Brain Connectome, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Franck Oury
- INSERM U1151, Institut Necker Enfants-Malades (INEM), Université Paris Descartes-Sorbonne, Paris Cité, Paris, France.
| | - Patricia Ducy
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| | - Vijay K Yadav
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Brain Connectome, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
| | - Gerard Karsenty
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
44
|
Alrumaih S, Alshibani N, Alssum L, Alshehri FA, AlMayrifi MA, AlMayouf M, Alrahlah A, Bautista LSJ. The impact of Resolvin E1 on bone regeneration in critical-sized calvarial defects of rat model-A gene expression and micro-CT analysis. J Periodontal Res 2024; 59:195-203. [PMID: 37947141 DOI: 10.1111/jre.13206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/15/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To investigate, in vivo, the effect of local application of Resolvin E1 (RvE1) on the bone regeneration of critical-size defects (CSDs) in Wistar rats utilizing gene expression and micro-computed tomographic (micro-CT) analysis. BACKGROUND The inflammation-resolving actions of RvE1 are well established. The molecular mechanism of its bone-regenerative actions has been of significant interest in recent years; however, there is limited information regarding the same. MATERIALS AND METHODS Thirty Wistar rats with a 5 mm induced critical-size calvarial defect were randomly allocated into four groups: no treatment/negative control (n = 5), treatment using bovine bone grafts/positive control (n = 5), treatment using local delivery of RvE1 (n = 11) and treatment using RvE1 mixed with bovine bone graft (n = 9). After 4 weeks, RNA isolation, complementary DNA synthesis and real-time polymerase chain reaction were used for genetic expression of alkaline phosphatase (ALP), osteocalcin (OCN) and osteopontin (OPN). The rats were sacrificed after 12 weeks and micro-CT imaging was performed to analyse the characteristics of the newly formed bone (NFB). The data were analysed using ANOVA and the least significant difference tests (α ≤ .05). RESULTS The RvE1 + bovine graft group had statistically highest mean NFB (20.75 ± 2.67 mm3 ) compared to other groups (p < .001). Similarly, RvE1 + bovine graft group also demonstrated statistically highest mean genetic expression of ALP (31.71 ± 2.97; p = .008) and OPN (34.78 ± 3.62; p < .001) compared to negative control and RvE1 groups. CONCLUSION Resolvin E1 with adjunct bovine bone graft demonstrated an enhanced bone regeneration compared to RvE1 or bovine graft alone in the calvarial defect of Wistar rats.
Collapse
Affiliation(s)
- Sara Alrumaih
- Department of Periodontics and Community Dentistry, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
- Hail Health Cluster, Ministry of Health, Hail, Saudi Arabia
| | - Nouf Alshibani
- Department of Periodontics and Community Dentistry, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Lamees Alssum
- Department of Periodontics and Community Dentistry, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Fahad A Alshehri
- Department of Periodontics and Community Dentistry, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed A AlMayrifi
- Prince Naif bin Abdulaziz Center for Health Research, Experimental Surgery and Animal Laboratory, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed AlMayouf
- General Director of Medical Services, Al Nakheel Center, Riyadh, Saudi Arabia
| | - Ali Alrahlah
- Restorative Dental Sciences Department, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
- Engineer Abdullah Bugshan Research Chair for Dental and Oral Rehabilitation, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Leonel S J Bautista
- Engineer Abdullah Bugshan Research Chair for Dental and Oral Rehabilitation, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Tavakol M, Liu J, Hoff SE, Zhu C, Heinz H. Osteocalcin: Promoter or Inhibitor of Hydroxyapatite Growth? LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:1747-1760. [PMID: 38181199 DOI: 10.1021/acs.langmuir.3c02948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2024]
Abstract
Osteocalcin is the most abundant noncollagenous bone protein and the functions in bone remineralization as well as in inhibition of bone growth have remained unclear. In this contribution, we explain the dual role of osteocalcin in the nucleation of new calcium phosphate during bone remodeling and in the inhibition of hydroxyapatite crystal growth at the molecular scale. The mechanism was derived using pH-resolved all-atom models for the protein, phosphate species, and hydroxyapatite, along with molecular dynamics simulations and experimental and clinical observations. Osteocalcin binds to (hkl) hydroxyapatite surfaces through multiple residues, identified in this work, and the fingerprint of binding residues varies as a function of the (hkl) crystal facet and pH value. On balance, the affinity of osteocalcin to hydroxyapatite slows down crystal growth. The unique tricalcium γ-carboxylglutamic acid (Gla) domain hereby rarely adsorbs to hydroxyapatite surfaces and faces instead toward the solution. The Gla domain enables prenucleation of calcium phosphate for new bone formation at a slightly acidic pH of 5. The growth of prenucleation clusters of calcium phosphate continues upon increase in pH value from 5 to 7 and is much less favorable, or not observed, on the native osteocalcin structure at and above neutral pH values of 7. The results provide mechanistic insight into the early stages of bone remodeling from the molecular scale, help inform mutations of osteocalcin to modify binding to apatites, support drug design, and guide toward potential cures for osteoporosis and hyperosteogeny.
Collapse
Affiliation(s)
- Mahdi Tavakol
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, Colorado 80301, United States
- Department of Mechanical Engineering, Sharif University of Technology, PO Box 11365-11155, Tehran, Iran
| | - Juan Liu
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, Colorado 80301, United States
| | - Samuel E Hoff
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, Colorado 80301, United States
| | - Cheng Zhu
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, Colorado 80301, United States
| | - Hendrik Heinz
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Ave, Boulder, Colorado 80301, United States
- Materials Science and Engineering Program, University of Colorado Boulder, 3415 Colorado Ave, Boulder, Colorado 80301, United States
| |
Collapse
|
46
|
Fusaro M, Barbuto S, Gallieni M, Cossettini A, Re Sartò GV, Cosmai L, Cianciolo G, La Manna G, Nickolas T, Ferrari S, Bover J, Haarhaus M, Marino C, Mereu MC, Ravera M, Plebani M, Zaninotto M, Cozzolino M, Bianchi S, Messa P, Gregorini M, Gasperoni L, Agosto C, Aghi A, Tripepi G. Real-world usage of Chronic Kidney Disease - Mineral Bone Disorder (CKD-MBD) biomarkers in nephrology practices. Clin Kidney J 2024; 17:sfad290. [PMID: 38223338 PMCID: PMC10784916 DOI: 10.1093/ckj/sfad290] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Indexed: 01/16/2024] Open
Abstract
Background Chronic kidney disease mineral bone disorder (CKD-MBD) is a condition characterized by alterations of calcium, phosphate, parathyroid hormone (PTH), and fibroblast growth factor 23 (FGF-23) metabolism that in turn promote bone disorders, vascular calcifications, and increase cardiovascular (CV) risk. Nephrologists' awareness of diagnostic, prognostic, and therapeutic tools to manage CKD-MBD plays a primary role in adequately preventing and managing this condition in clinical practice. Methods A national survey (composed of 15 closed questions) was launched to inquire about the use of bone biomarkers in the management of CKD-MBD patients by nephrologists and to gain knowledge about the implementation of guideline recommendations in clinical practice. Results One hundred and six Italian nephrologists participated in the survey for an overall response rate of about 10%. Nephrologists indicated that the laboratories of their hospitals were able to satisfy request of ionized calcium levels, 105 (99.1%) of both PTH and alkaline phosphatase (ALP), 100 (94.3%) of 25(OH)D, and 61 (57.5%) of 1.25(OH)2D; while most laboratories did not support the requests of biomarkers such as FGF-23 (intact: 88.7% and c-terminal: 93.4%), Klotho (95.3%; soluble form: 97.2%), tartrate-resistant acid phosphatase 5b (TRAP-5b) (92.5%), C-terminal telopeptide (CTX) (71.7%), and pro-collagen type 1 N-terminal pro-peptide (P1NP) (88.7%). As interesting data regarding Italian nephrologists' behavior to start treatment of secondary hyperparathyroidism (sHPT), the majority of clinicians used KDOQI guidelines (n = 55, 51.9%). In contrast, only 40 nephrologists (37.7%) relied on KDIGO guidelines, which recommended referring to values of PTH between two and nine times the upper limit of the normal range. Conclusion Results point out a marked heterogeneity in the management of CKD-MBD by clinicians as well as a suboptimal implementation of guidelines in Italian clinical practice.
Collapse
Affiliation(s)
- Maria Fusaro
- National Research Council (CNR), Institute of Clinical Physiology (IFC), Pisa, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Simona Barbuto
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Maurizio Gallieni
- Department of Biomedical and Clinical Sciences ‘Luigi Sacco’, Università di Milano, Milano, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milano, Milano, Italy
- Division of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Fatebenefratelli-Sacco, Fatebenefratelli Hospital, Milan, Italy
| | - Althea Cossettini
- Post-Graduate School of Specialization in Nephrology, University of Milano, Milano, Italy
| | | | - Laura Cosmai
- Division of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Fatebenefratelli-Sacco, Fatebenefratelli Hospital, Milan, Italy
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Thomas Nickolas
- Department of Medicine, Division of Nephrology, Columbia University, New York, NY, USA
| | - Serge Ferrari
- Service des Maladies Osseuses, Département de Médecine, HUG, Geneva, Switzerland
| | - Jordi Bover
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, Badalona (Barcelona), Spain
| | - Mathias Haarhaus
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Carmela Marino
- National Research Council (CNR), Institute of Clinical Physiology (IFC), Reggio Calabria, Italy
| | | | - Maura Ravera
- Nephrology, Dialysis, and Transplantation, University of Genoa and Policlinico San Martino, 16132 Genoa, Italy
| | - Mario Plebani
- Laboratory Medicine Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Martina Zaninotto
- Laboratory Medicine Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Mario Cozzolino
- Department of Health Sciences, Renal Division, University of Milan, ASST Santi Paolo e Carlo, Milan, Italy
| | - Stefano Bianchi
- Department of Internal Medicine, Nephrology and Dialysis Complex Operative Unit, Livorno, Italy
| | - Piergiorgio Messa
- Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Lorenzo Gasperoni
- Nephrology and Dialysis Unit, Infermi Hospital, AUSL Romagna, Rimini, Italy
| | - Caterina Agosto
- Pediatric Pain and Palliative Care Service, Department of Women's and Children's Health, Padua University Hospital, Padua, Italy
| | | | - Giovanni Tripepi
- National Research Council (CNR), Institute of Clinical Physiology (IFC), Reggio Calabria, Italy
| |
Collapse
|
47
|
Alake SE, Ice J, Robinson K, Price P, Hatter B, Wozniak K, Lin D, Chowanadisai W, Smith BJ, Lucas EA. Reduced estrogen signaling contributes to bone loss and cardiac dysfunction in interleukin-10 knockout mice. Physiol Rep 2024; 12:e15914. [PMID: 38217044 PMCID: PMC10787104 DOI: 10.14814/phy2.15914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
Characterization of the interleukin (IL)-10 knockout (KO) mouse with chronic gut inflammation, cardiovascular dysfunction, and bone loss suggests a critical role for this cytokine in interorgan communication within the gut, bone, and cardiovascular axis. We sought to understand the role of IL-10 in the cross-talk between these systems. Six-week-old IL-10 KO mice and their wild type (WT) counterparts were maintained on a standard rodent diet for 3 or 6 months. Gene expression of proinflammatory markers and Fgf23, serum 17β-estradiol (E2), and cardiac protein expression were assessed. Ileal Il17a and Tnf mRNA increased while Il6 mRNA increased in the bone and heart by at least 2-fold in IL-10 KO mice. Bone Dmp1 and Phex mRNA were repressed at 6 months in IL-10 KO mice, resulting in increased Fgf23 mRNA (~4-fold) that contributed to increased fibrosis. In the IL-10 KO mice, gut bacterial β-glucuronidase activity and ovarian Cyp19a1 mRNA were lower (p < 0.05), consistent with reduced serum E2 and reduced cardiac pNOS3 (Ser1119 ) in these mice. Treatment of ileal lymphocytes with E2 reduced gut inflammation in WT but not IL-10 KO mice. In conclusion, our data suggest that diminished estrogen and defective bone mineralization increased FGF23 which contributed to cardiac fibrosis in the IL-10 KO mouse.
Collapse
Affiliation(s)
- Sanmi E. Alake
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - John Ice
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Kara Robinson
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Payton Price
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Bethany Hatter
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Karen Wozniak
- Department of Microbiology and Molecular GeneticsOklahoma State UniversityStillwaterOklahomaUSA
| | - Dingbo Lin
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Winyoo Chowanadisai
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Brenda J. Smith
- Department of Obstetrics and GynecologyIndiana School of MedicineIndianapolisIndianaUSA
- Indiana Center for Musculoskeletal HealthIndiana School of MedicineIndianapolisIndianaUSA
| | - Edralin A. Lucas
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| |
Collapse
|
48
|
da Silva Sasso GR, Florencio-Silva R, de Pizzol-Júnior JP, Gil CD, Simões MDJ, Sasso-Cerri E, Cerri PS. Additional Insights Into the Role of Osteocalcin in Osteoblast Differentiation and in the Early Steps of Developing Alveolar Process of Rat Molars. J Histochem Cytochem 2023; 71:689-708. [PMID: 37953508 PMCID: PMC10691409 DOI: 10.1369/00221554231211630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/05/2023] [Indexed: 11/14/2023] Open
Abstract
This study investigated whether osteocalcin (OCN) is present in osteoblast precursors and its relationship with initial phases of alveolar process formation. Samples of maxillae of 16-, 18-, and 20-day-old rat embryos (E16, E18, and E20, respectively), and 05-, 10-, and 15-day-old postnatal rats (P05, P10, and P15, respectively) were fixed and embedded in paraffin or araldite. Immunohistochemistry for osterix (Osx), alkaline phosphatase (ALP), and OCN detection was performed and the number of immunolabelled cells was computed. Non-decalcified sections were subjected to the von Kossa method combined with immunohistochemistry for Osx or OCN detection. For OCN immunolocalization, samples were fixed in 0.5% glutaraldehyde/2% formaldehyde and embedded in LR White resin. The highest number of ALP- and OCN-immunolabelled cells was observed in dental follicle of E16 specimens, mainly in basal portions of dental alveolus. In corresponding regions, osteoblasts in differentiation adjacent to von Kossa-positive bone matrix exhibited Osx and OCN immunoreactivity. Ultrastructural analysis revealed OCN immunoreactive particles inside osteoblast in differentiation, and in bone matrix associated with collagen fibrils and within matrix vesicles, at early stages of alveolar process formation. Our results indicate that OCN plays a role in osteoblast differentiation and may regulate calcium/phosphate precipitation during early mineralization of the alveolar process.
Collapse
Affiliation(s)
- Gisela Rodrigues da Silva Sasso
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brasil
| | - Rinaldo Florencio-Silva
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brasil
- Departamento de Ginecologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brasil
| | - José Paulo de Pizzol-Júnior
- Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Cristiane Damas Gil
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brasil
| | - Manuel de Jesus Simões
- Disciplina de Histologia e Biologia Estrutural, Departamento de Morfologia e Genética, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brasil
| | - Estela Sasso-Cerri
- Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Paulo Sérgio Cerri
- Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
49
|
Bian X, Jin L, Wang Y, Yuan M, Yao Z, Ning B, Gao W, Guo C. Riboflavin deficiency reduces bone mineral density in rats by compromising osteoblast function. J Nutr Biochem 2023; 122:109453. [PMID: 37788723 DOI: 10.1016/j.jnutbio.2023.109453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/05/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023]
Abstract
Insufficient riboflavin intake has been associated with poor bone health. This study aimed to investigate the effect of riboflavin deficiency on bone health in vivo and in vitro. Riboflavin deficiency was successfully developed in rats and osteoblasts. The results indicated that bone mineral density, serum bone alkaline phosphatase, bone phosphorus, and bone calcium were significantly decreased while serum ionized calcium and osteocalcin were significantly increased in the riboflavin-deficient rats. Riboflavin deficiency also induced the reduction of Runx2, Osterix, and BMP-2/Smad1/5/9 cascade in the femur. These results were further verified in cellular experiments. Our findings demonstrated that alkaline phosphatase activities and calcified nodules were significantly decreased while intracellular osteocalcin and pro-collagen I c-terminal propeptide were significantly increased in the riboflavin-deficient osteoblasts. Additionally, the protein expression of Osterix, Runx2, and BMP-2/Smad1/5/9 cascade were significantly decreased while the protein expression of p-p38 MAPK were significantly increased in the riboflavin-deficient cells compared to the control cells. Blockage of p38 MAPK signaling pathway with SB203580 reversed these effects in riboflavin-deficient osteoblastic cells. Our data suggest that riboflavin deficiency causes osteoblast malfunction and retards bone matrix mineralization via p38 MAPK/BMP-2/Smad1/5/9 signaling pathway.
Collapse
Affiliation(s)
- Xiangyu Bian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Lu Jin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Yanxian Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Man Yuan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Zhanxin Yao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Baoan Ning
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Weina Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China.
| | - Changjiang Guo
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, PR China.
| |
Collapse
|
50
|
Tian Y, Zhao L, Gui Z, Liu S, Liu C, Yu T, Zhang L. PI3K/AKT signaling activates HIF1α to modulate the biological effects of invasive breast cancer with microcalcification. NPJ Breast Cancer 2023; 9:93. [PMID: 37957150 PMCID: PMC10643473 DOI: 10.1038/s41523-023-00598-z] [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: 02/10/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Microcalcification (MC) is a valuable diagnostic indicator of breast cancer, and it is reported to be associated with increased tumor aggressiveness and poor prognosis. Nevertheless, the exact potential molecular mechanism is not completely understood. Here, we find that the mineralized invasive breast cancer (IBC) cells not only increased their proliferation and migration, but also showed the characteristic of doxorubicin resistance. The PI3K/AKT signaling pathway is associated with the generation of calcification in IBC, and it activates the transcription and translation of its downstream hypoxia-inducible factor 1α (HIF1α). Knockdown of HIF1α protein significantly downregulated cell proliferation and migration while calcification persists. Meanwhile, calcified breast cancer cells restored sensitivity to doxorubicin because of suppressed HIF1α expression. In addition, we provide initial data on the underlying value of HIF1α as a biomarker of doxorubicin resistance. These findings provide a new direction for exploring microcalcifications in IBC.
Collapse
Affiliation(s)
- Yao Tian
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Lu Zhao
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Zhengwei Gui
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Chenguang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Tianyao Yu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Lin Zhang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China.
| |
Collapse
|