1
|
Weng K, He Y, Weng X, Yuan Y. Exercise alleviates osteoporosis by regulating the secretion of the Senescent Associated Secretory Phenotype. Bone 2025; 196:117485. [PMID: 40216288 DOI: 10.1016/j.bone.2025.117485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
As the elderly population grows, the number of patients with metabolic bone diseases such as osteoporosis has increased sharply, posing a significant threat to public health and social economics. Although pharmacological therapies for osteoporosis demonstrate therapeutic benefits, their prolonged use is associated with varying degrees of adverse effects. As a non-pharmacological intervention, exercise is widely recognized for its cost-effectiveness, safety, and lack of toxic side effects, making it a recommended treatment for osteoporosis prevention and management. Previous studies have demonstrated that exercise can improve metabolic bone diseases by modulating the Senescent Associated Secretory Phenotype (SASP). However, the mechanisms through which exercise influences SASP remain unclear. Therefore, this review aims to summarize the effects of exercise on SASP and elucidate the specific mechanisms by which exercise regulates SASP to alleviate osteoporosis, providing a theoretical basis for osteoporosis through exercise and developing targeted therapies.
Collapse
Affiliation(s)
- Kaihong Weng
- Graduate School, Guangzhou Sport University, 510500 Guangzhou, China
| | - Yuting He
- Graduate School, Guangzhou Sport University, 510500 Guangzhou, China
| | - Xiquan Weng
- School of Exercise and Health, Guangzhou Sport University, 510500 Guangzhou, China; Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, 510500 Guangzhou, China.
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, 510500 Guangzhou, China; Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, 510500 Guangzhou, China.
| |
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
|
Kinoshita S, Onuma S, Yamazaki N, Shibukawa Y, Ozono K, Michigami T, Kawai M. Lack of PTEN in Osteocytes Increases Lipocalin-2 Level and Confers Resistance to High-Fat Diet-Induced Obesity in Mice. Endocrinology 2025; 166:bqaf026. [PMID: 39921437 DOI: 10.1210/endocr/bqaf026] [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: 10/22/2024] [Revised: 12/25/2024] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Osteocytes have been shown to play critical roles in the regulation of a wide range of metabolic processes. However, their role in the regulation of glucose metabolism remains to be determined despite accumulating evidence of the integral role of osteoblasts in this regulation, in which osteoblast-derived lipocalin-2 (LCN2) has been shown to regulate glucose metabolism. Additionally, Lcn2 expression is induced by AKT activation. These results led us to hypothesize that AKT activation in osteocytes regulates glucose metabolism by modulating Lcn2 expression. Therefore, in this study, the Pten gene was deleted in osteocytes to activate AKT signaling by crossing Pten-flox mice with Dmp1-Cre mice (PtenOcy-/- mice). Deleting Pten expression in osteocytes resulted in osteocytic AKT activation, which was associated with decreased adipose tissue mass and enhanced insulin sensitivity. Expression of Pparg2 and lipogenesis-associated genes were decreased in the adipose tissue of PtenOcy-/- mice. Mechanistically, the lack of Phosphatase and Tensin Homolog Deleted from Chromosome 10 (PTEN) in osteocytes increased Lcn2 expression in the femur, which was associated with increased serum and urine LCN2 levels. The urinary LCN2 level was negatively associated with white adipose tissue mass. Additionally, the treatment of primary white adipocytes with recombinant LCN2 reduced the expression of Pparg2 and lipogenesis-related genes. These results suggest that the absence of PTEN in osteocytes increases the expression of Lcn2, which acts in the adipose tissue to suppress lipogenesis, resulting in enhanced insulin sensitivity in these mice. This study provides novel insights into the critical role of AKT activation in osteocytes in regulating glucose metabolism by increasing Lcn2 expression.
Collapse
Affiliation(s)
- Saori Kinoshita
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
| | - Shinsuke Onuma
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
| | - Natsuko Yamazaki
- Department of Molecular Genetics and Endocrinology, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
| | - Yukinao Shibukawa
- Department of Molecular Genetics and Endocrinology, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
| | - Keiichi Ozono
- Center for Promoting Treatment of Intractable Diseases, ISEIKAI International General Hospital, Osaka 530-0052, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
- Department of Molecular Genetics and Endocrinology, Research Institute, Osaka Women's and Children's Hospital, 594-1101, Osaka, Japan
| |
Collapse
|
4
|
Sentosa DD, Metcalfe RD, Sims NA, Putoczki TL, Griffin MDW. The structure of the IL-11 signalling complex provides insight into receptor variants associated with craniosynostosis. FEBS J 2025; 292:500-509. [PMID: 39462650 DOI: 10.1111/febs.17307] [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: 08/04/2024] [Revised: 09/25/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
Interleukin 11 (IL-11), a member of the IL-6 family of cytokines, has roles in haematopoiesis, inflammation, bone metabolism, and craniofacial development. IL-11 also has pathological roles in chronic inflammatory diseases, fibrosis, and cancer. In this structural snapshot, we explore our recently published cryo-EM structure of the human IL-11 signalling complex to understand the molecular mechanisms of complex formation and disease-associated mutations. IL-11 signals by binding to its cell surface receptors, the IL-11 receptor α subunit (IL-11Rα) and glycoprotein 130 (gp130), to form a hexameric signalling complex. We examine the locations within the complex of receptor sequence variants that are associated with craniosynostosis and craniosynostosis-like phenotypes and speculate on potential molecular mechanisms leading to defects in signalling function. While these causative amino acid sequence changes in IL-11Rα are generally distal to interfaces between components of the complex, important structural residues are highly represented, including proline residues, cysteine residues involved in disulfide bonds, and residues within or surrounding the tryptophan-arginine ladder. We also note the locations and potential effects of amino acid substitutions within the extracellular domains of gp130 that are associated with craniosynostosis. As focus on the physiological and pathological functions of IL-11 grows, the importance of high-resolution structural knowledge of IL-11 signalling to understand disease-associated mutations and to inform therapeutic strategies will only increase.
Collapse
Affiliation(s)
- Darlene D Sentosa
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
| | - Riley D Metcalfe
- Centre for Structural Biology, Centre for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, Australia
- Mary Mackillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Tracy L Putoczki
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Department of Surgery, University of Melbourne, Parkville, Australia
| | - Michael D W Griffin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
| |
Collapse
|
5
|
Lokau J, Garbers Y, Vicente MM, Dittrich A, Meltendorf S, Lingel H, Münster-Kühnel AK, Brunner-Weinzierl M, Garbers C. Long-term increase in soluble interleukin-6 receptor levels in convalescents after mild COVID-19 infection. Front Immunol 2025; 15:1488745. [PMID: 39835136 PMCID: PMC11743636 DOI: 10.3389/fimmu.2024.1488745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Serum levels of interleukin-6 (IL-6) are increased in COVID-19 patients. IL-6 is an effective therapeutic target in inflammatory diseases and tocilizumab, a monoclonal antibody that blocks signaling via the IL-6 receptor (IL-6R), is used to treat patients with severe COVID-19. However, the IL-6R exists in membrane-bound and soluble forms (sIL-6R), and the sIL-6R in combination with soluble glycoprotein 130 (sgp130) forms an IL-6-neutralizing buffer system capable of neutralizing small amounts of IL-6. Methods In this study, we analyzed serum levels of IL-6, sIL-6R and sgp130 in the serum of COVID-19 convalescent individuals with a history of mild COVID-19 disease and in acute severely ill COVID-19 patients compared to uninfected control subjects. Furthermore, we used single cell RNA sequencing data in order to determine which immune cell types are sources and targets of the individual cytokines and whether their expression is altered in severe COVID-19 patients. Results We find that sIL-6R levels are not only increased in acute severely ill patients, but also in convalescents after a mild COVID-19 infection. We show that this increase in sIL-6R results in an enhanced capacity of the sIL-6R/sgp130 buffer system, but that significantly enhanced free IL-6 is still present due to an overload of the buffer. Further, we identify IL-6 serum levels, age and the number of known pre-existing medical conditions as crucial determinants of disease outcome for the patients. We also show that IL-11 has no major systemic role in COVID-19 patients and that sCD25 is only increased in acute severely ill COVID-19 patients, but not in mild convalescent individuals. Discussion In conclusion, our study shows long-lasting alterations of the IL-6 system after COVID-19 disease, which might be relevant when applying anti-IL-6 or anti-IL-6R therapy.
Collapse
Affiliation(s)
- Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Yvonne Garbers
- Faculty of Management, Culture and Technology (Lingen campus), Osnabrück University of Applied Sciences, Lingen, Germany
| | - Manuel M. Vicente
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Anna Dittrich
- Department of Systems Biology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Stefan Meltendorf
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | - Monika Brunner-Weinzierl
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| |
Collapse
|
6
|
Mathavan N, Singh A, Marques FC, Günther D, Kuhn GA, Wehrle E, Müller R. Spatial transcriptomics in bone mechanomics: Exploring the mechanoregulation of fracture healing in the era of spatial omics. SCIENCE ADVANCES 2025; 11:eadp8496. [PMID: 39742473 DOI: 10.1126/sciadv.adp8496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
In recent decades, the field of bone mechanobiology has sought experimental techniques to unravel the molecular mechanisms governing the phenomenon of mechanically regulated fracture healing. Each cell within a fracture site resides within different local microenvironments characterized by different levels of mechanical strain; thus, preserving the spatial location of each cell is critical in relating cellular responses to mechanical stimuli. Our spatial transcriptomics-based "mechanomics" platform facilitates spatially resolved analysis of the molecular profiles of cells with respect to their local in vivo mechanical environment by integrating time-lapsed in vivo micro-computed tomography, spatial transcriptomics, and micro-finite element analysis. We investigate the transcriptomic responses of cells as a function of the local strain magnitude by identifying the differential expression of genes in regions of high and low strain within a fracture site. Our platform thus has the potential to address fundamental open questions within the field and to discover mechano-responsive targets to enhance fracture healing.
Collapse
Affiliation(s)
| | - Amit Singh
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | | | - Denise Günther
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
- AO Research Institute Davos, Davos Platz, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
7
|
Yu C, Sun R, Yang W, Gu T, Ying X, Ye L, Zheng Y, Fan S, Zeng X, Yao S. Exercise ameliorates osteopenia in mice via intestinal microbial-mediated bile acid metabolism pathway. Theranostics 2025; 15:1741-1759. [PMID: 39897551 PMCID: PMC11780523 DOI: 10.7150/thno.104186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/09/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Physical exercise is essential for skeletal integrity and bone health. The gut microbiome, as a pivotal modulator of overall physiologic states, is closely associated with skeletal homeostasis and bone metabolism. However, the potential role of intestinal microbiota in the exercise-mediated bone gain remains unclear. Methods: We conducted microbiota depletion and fecal microbiota transplantation (FMT) in ovariectomy (OVX) mice and aged mice to investigate whether the transfer of gut ecological traits could confer the exercise-induced bone protective effects. The study analyzed the gut microbiota and metabolic profiles via 16S rRNA gene sequencing and LC-MS untargeted metabolomics to identify key microbial communities and metabolites responsible for bone protection. Transcriptome sequencing and RNA interference were employed to explore the molecular mechanisms. Results: We found that gut microbiota depletion hindered the osteogenic benefits of exercise, and FMT from exercised osteoporotic mice effectively mitigated osteopenia. Comprehensive profiling of the microbiome and metabolome revealed that the exercise-matched FMT reshaped intestinal microecology and metabolic landscape. Notably, alterations in bile acid metabolism, specifically the enrichment of taurine and ursodeoxycholic acid, mediated the protective effects on bone mass. Mechanistically, FMT from exercised mice activated the apelin signaling pathway and restored the bone-fat balance in recipient MSCs. Conclusion: Our study underscored the important role of the microbiota-metabolic axis in the exercise-mediated bone gain, heralding a potential breakthrough in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Rongtai Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Wentao Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Xiaozhang Ying
- Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang 310016, China
| | - Lin Ye
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Yang Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
- Research Institute of Orthopedics, The Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Shasha Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration, Translational Research of Zhejiang Province Hangzhou, Zhejiang 310016, China
| |
Collapse
|
8
|
Han Y, Gao H, Gao J, Yang Y, He C. Low-intensity pulsed ultrasound regulates bone marrow mesenchymal stromal cells differentiation and inhibits bone loss by activating the IL-11-Wnt/β-catenin signaling pathway. Int Immunopharmacol 2024; 143:113380. [PMID: 39405933 DOI: 10.1016/j.intimp.2024.113380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Osteoporosis (OP) is a common metabolic bone disease. Low-intensity pulsed ultrasound (LIPUS) can effectively promote bone formation and fracture healing. The Wnt/β-catenin signaling pathway is crucial for regulating bone homeostasis and bone diseases, and its downregulation is one of the main mechanisms of osteoporosis pathogenesis. Interleukin-11 (IL-11), which is regulated by mechanical stress, is a key factor in bone remodeling. Here, we investigated the optimal intervention parameters for LIPUS, the relationships among LIPUS, IL-11, and the Wnt/β-catenin signaling pathway, and the effects of LIPUS on bone loss and potential molecular mechanisms in ovariectomized (OVX) mice. METHODS Bone marrow mesenchymal stromal cells (BMSCs) were subjected to LIPUS intervention for 0, 10, or 20 min to determine the optimal intervention time. The mediating role of IL-11 in LIPUS intervention was explored through IL-11 knockdown and overexpression. Finally, animal experiments were conducted to investigate the in vivo therapeutic effects of LIPUS. RESULTS The optimal intervention time for LIPUS was 20 min. LIPUS promoted IL-11 expression and upregulated the Wnt/β-catenin signaling pathway, thereby promoting osteogenic differentiation and inhibiting adipogenic differentiation of BMSCs. IL-11 mediates the regulation of the Wnt/β-catenin signaling pathway by LIPUS. Additionally, LIPUS effectively improved the bone microstructure in ovariectomized mice, inhibited bone loss, promoted IL-11 expression in bone tissue, and activated the Wnt/β-catenin signaling pathway in the femur. CONCLUSION Low-intensity pulsed ultrasound can regulate BMSCs differentiation and inhibit bone loss by promoting IL-11 expression and activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yijing Han
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000
| | - Hui Gao
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000
| | - Jing Gao
- LIFU Medical Research Center, Sichuan Taiyou Technology Co., Ltd., Chengdu, Sichuan, China, 610000
| | - Yonghong Yang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000.
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000.
| |
Collapse
|
9
|
Liu Z, Lin Z, Chen Y, Lu M, Hong W, Yu B, Liu G. Lipoteichoic Acid Rescued Age-Related Bone Loss by Enhancing Neuroendocrine and Growth Hormone Secretion Through TLR2/COX2/PGE2 Signalling Pathway. J Cell Mol Med 2024; 28:e70247. [PMID: 39622781 PMCID: PMC11611525 DOI: 10.1111/jcmm.70247] [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] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
The phenomenon of brain-bone crosstalk pertains to the intricate interaction and communication pathways between the central nervous system and the skeletal system. Disruption in brain-bone crosstalk, particularly in disorders such as osteoporosis, can result in skeletal irregularities. Consequently, investigating and comprehending this communication network holds paramount importance in the realm of bone disease prevention and management. In this study, we found that Staphylococcus aureus lipoteichoic acid promoted the conversion of arachidonic acid to PGE2 by interacting with TLR2 receptors acting on the surface of microglial cells in the pituitary gland, leading to the upregulation of COX-2 expression. Subsequently, PGE2 bound to the EP4 receptor of growth hormone-secreting cells and activated the intracellular CREB signalling pathway, promoting GH secretion and ameliorating age-related bone loss.
Collapse
Affiliation(s)
- Zixian Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- The Second Hospital and Clinical Medical SchoolLanzhou UniversityLanzhouChina
| | - Zexin Lin
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yingqi Chen
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Mincheng Lu
- Department of Orthopedic, Shenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Weisheng Hong
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Bin Yu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guanqiao Liu
- Department of Orthopaedics, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
10
|
Lee SJ, Jung DK, Im S, You C, Kim JE, Bae JS, Kim MS, Yea K, Park EK. Ank-mediated pyrophosphate regulates shear stress-induced small extracellular vesicle production in 3D-cultured osteocytes. Anim Cells Syst (Seoul) 2024; 28:495-505. [PMID: 39445097 PMCID: PMC11497576 DOI: 10.1080/19768354.2024.2409460] [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: 06/27/2024] [Revised: 08/20/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Osteocytes are located in the lacunae of fluid-filled bone and communicate with neighboring or distant cells by secreting small extracellular vesicles (sEVs) and growth factors as well as via dendrite-dendrite direct connections. However, the mechanism regulating sEV production in osteocytes is yet to be elucidated. In this study, we investigated sEV production and its underlying mechanism in osteocytes cultured on a three dimensional (3D) scaffold. We employed a perfusion system to apply shear stress stimulation to MLO-Y4 cells cultured on a 3D biphasic calcium phosphate (BCP) scaffold and analyzed sEV production and gene expression using RNA sequencing. We found that the expression of genes associated with sEV biogenesis and the secretory pathway were enhanced by fluid shear stress in MLO-Y4 cells cultured on a 3D BCP scaffold. In particular, fluid shear stress induced the expression of Ank, a pyrophosphate transporter, in 3D-cultured MLO-Y4 cells. The role of Ank in sEV production was further examined. Probenecid, an Ank inhibitor, significantly suppressed shear stress-induced sEV production, whereas Ank cDNA overexpression stimulated it. The inhibition of shear stress-induced sEV production by probenecid was recovered by the exogenous addition of pyrophosphate to MLO-Y4 cells. These findings suggest that shear stress-mediated sEV production in 3D-cultured osteocytes is regulated by extracellular pyrophosphate transported by Ank.
Collapse
Affiliation(s)
- Su Jeong Lee
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu, Republic of Korea
| | - Deuk Kju Jung
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu, Republic of Korea
| | - Soomin Im
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu, Republic of Korea
| | - Changkook You
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu, Republic of Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute (CMRI), School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Mee-seon Kim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu, Republic of Korea
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
11
|
Cheng TY, Wu WT, Peng CH, Liu KL, Yao TK, Yu TC, Chen IH, Yeh KT. Effect of aerobic exercise on bone health in postmenopausal women with obesity: Balancing benefits with caloric restriction and resistance exercise. Tzu Chi Med J 2024; 36:377-386. [PMID: 39421495 PMCID: PMC11483090 DOI: 10.4103/tcmj.tcmj_40_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/01/2024] [Accepted: 05/21/2024] [Indexed: 10/19/2024] Open
Abstract
The decline in bone mineral density (BMD) poses a significant concern for postmenopausal women with obesity. Research indicates that aerobic exercises show potential for enhancing bone health. However, there remains no consensus regarding their effects on BMD. This study aimed to evaluate the effect of various exercise interventions on BMD and overall health among postmenopausal women with obesity, with particular attention to caloric restriction (CR). Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 guidelines, we performed a comprehensive literature search on PubMed, targeting studies published up to August 2023. Our search focused on aerobic exercise, resistance training, and combined exercise modalities, examining their impact on BMD, body composition, and physical fitness in postmenopausal women with obesity. We reviewed 11 studies, predominantly on aerobic exercise, involving women who are overweight and sedentary, nine of which were randomized trials. Our findings suggest that aerobic exercise has a mild protective effect on BMD and can significantly reduce fat mass. Notably, when combined with CR, aerobic exercise not only enhances the reduction of fat tissue mass but also potentially offers a certain level of protection for BMD. Additionally, the intervention combining aerobic exercise with resistance training emerges as a key promoter of bone health, underscoring the importance of tailored exercise programs for this population. Consequently, balanced dietary patterns (like the Mediterranean diet), combined with exercise, are recommended for optimal health outcomes. Tailored exercise programs integrating both aerobic and resistance training are crucial for sustaining overall health and bone density in this population.
Collapse
Affiliation(s)
- Ti-Yen Cheng
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Tien Wu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Cheng-Huan Peng
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Kuan-Lin Liu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ting-Kuo Yao
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tzai-Chiu Yu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ing-Ho Chen
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Kuang-Ting Yeh
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Clinical Pharmacy, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
12
|
Hao R, Tang H, Ding C, Rajbanshi B, Liu Y, Ma D, Duan Z, Qi Y, Dai L, Zhang B, Zhang A, Zhang X. A Novel Piezo1 Agonist Promoting Mesenchymal Stem Cell Proliferation and Osteogenesis to Attenuate Disuse Osteoporosis. SMALL SCIENCE 2024; 4:2400061. [PMID: 40212079 PMCID: PMC11935128 DOI: 10.1002/smsc.202400061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/12/2024] [Indexed: 04/13/2025] Open
Abstract
Disuse osteoporosis (OP) is a state of bone loss due to lack of mechanical stimuli, probably induced by prolonged bed rest, neurological diseases, as well as microgravity. Currently the precise treatment strategies of disuse OP remain largely unexplored. Piezo1, a mechanosensitive calcium (Ca2+) ion channel, is a key force sensor mediating mechanotransduction and it is demonstrated to regulate bone homeostasis and osteogenesis in response to mechanical forces. Using structure-based drug design, a novel small-molecule Piezo1 agonist, MCB-22-174, which can effectively activate Piezo1 and initiate Ca2+ influx, is developed and is more potent than the canonical Piezo1 agonist, Yoda1. Moreover, MCB-22-174 is found as a safe Piezo1 agonist without any signs of serious toxicity. Mechanistically, Piezo1 activation promotes the proliferation of bone marrow mesenchymal stem cells by activating the Ca2+-related extracellular signal-related kinases and calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) pathway. Importantly, MCB-22-174 could effectively promote osteogenesis and attenuate disuse OP in vivo. Overall, the findings provide a promising therapeutic strategy for disuse OP by chemical activation of Piezo1.
Collapse
Affiliation(s)
- Ruihan Hao
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Hairong Tang
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Chunyong Ding
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Bhavana Rajbanshi
- Department of Dermatology and VenereologyTongji University School of MedicineShanghai200092China
| | - Yuhang Liu
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Ding Ma
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Zhouyi Duan
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Yuxin Qi
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and MinistryGuangxi Medical UniversityNanningGuangxi530021China
| | - Liming Dai
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| | - Bingjun Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| | - Ao Zhang
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Xiaoling Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| |
Collapse
|
13
|
Nomura K, Kinoshita S, Mizusaki N, Senga Y, Sasaki T, Kitamura T, Sakaue H, Emi A, Hosooka T, Matsuo M, Okamura H, Amo T, Wolf AM, Kamimura N, Ohta S, Itoh T, Hayashi Y, Kiyonari H, Krook A, Zierath JR, Kasuga M, Ogawa W. Adaptive gene expression of alternative splicing variants of PGC-1α regulates whole-body energy metabolism. Mol Metab 2024; 86:101968. [PMID: 38885788 PMCID: PMC11254180 DOI: 10.1016/j.molmet.2024.101968] [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: 03/07/2024] [Revised: 05/23/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
The transcriptional coactivator PGC-1α has been implicated in the regulation of multiple metabolic processes. However, the previously reported metabolic phenotypes of mice deficient in PGC-1α have been inconsistent. PGC-1α exists as multiple isoforms, including variants transcribed from an alternative first exon. We show here that alternative PGC-1α variants are the main entity that increases PGC-1α during exercise. These variants, unlike the canonical isoform of PGC-1α, are robustly upregulated in human skeletal muscle after exercise. Furthermore, the extent of this upregulation correlates with oxygen consumption. Mice lacking these variants manifest impaired energy expenditure during exercise, leading to the development of obesity and hyperinsulinemia. The alternative variants are also upregulated in brown adipose tissue in response to cold exposure, and mice lacking these variants are intolerant of a cold environment. Our findings thus indicate that an increase in PGC-1α expression, attributable mostly to upregulation of alternative variants, is pivotal for adaptive enhancement of energy expenditure and heat production and thereby essential for the regulation of whole-body energy metabolism.
Collapse
Affiliation(s)
- Kazuhiro Nomura
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Shinichi Kinoshita
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Nao Mizusaki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoko Senga
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Tsutomu Sasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan; Diabetes Therapeutics and Research Center, University of Tokushima, Tokushima 770-8503, Japan
| | - Aki Emi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Tetsuya Hosooka
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Masahiro Matsuo
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Hitoshi Okamura
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan; Department of Neuroscience, Graduate School of Medicine, Kyoto University, Kyoto 606-8303, Japan
| | - Taku Amo
- Department of Applied Chemistry, National Defense Academy, Yokosuka 239-8686, Japan
| | - Alexander M Wolf
- Department of Biochemistry and Cell Biology, Graduate School of Medicine, Nippon Medical School, Kawasaki 211-8533, Japan
| | - Naomi Kamimura
- Department of Biochemistry and Cell Biology, Graduate School of Medicine, Nippon Medical School, Kawasaki 211-8533, Japan; Laboratory for Clinical Research, Collaborative Research Center, Nippon Medical School, Tokyo 113-8602, Japan
| | - Shigeo Ohta
- Department of Biochemistry and Cell Biology, Graduate School of Medicine, Nippon Medical School, Kawasaki 211-8533, Japan
| | - Tomoo Itoh
- Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoshitake Hayashi
- Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Masato Kasuga
- The Institute of Medical Science, Asahi Life Foundation, Tokyo 100-0005, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| |
Collapse
|
14
|
Qiu M, Chang L, Tang G, Ye W, Xu Y, Tulufu N, Dan Z, Qi J, Deng L, Li C. Activation of the osteoblastic HIF-1α pathway partially alleviates the symptoms of STZ-induced type 1 diabetes mellitus via RegIIIγ. Exp Mol Med 2024; 56:1574-1590. [PMID: 38945950 PMCID: PMC11297314 DOI: 10.1038/s12276-024-01257-4] [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: 07/07/2023] [Revised: 02/04/2024] [Accepted: 03/19/2024] [Indexed: 07/02/2024] Open
Abstract
The hypoxia-inducible factor-1α (HIF-1α) pathway coordinates skeletal bone homeostasis and endocrine functions. Activation of the HIF-1α pathway increases glucose uptake by osteoblasts, which reduces blood glucose levels. However, it is unclear whether activating the HIF-1α pathway in osteoblasts can help normalize glucose metabolism under diabetic conditions through its endocrine function. In addition to increasing bone mass and reducing blood glucose levels, activating the HIF-1α pathway by specifically knocking out Von Hippel‒Lindau (Vhl) in osteoblasts partially alleviated the symptoms of streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM), including increased glucose clearance in the diabetic state, protection of pancreatic β cell from STZ-induced apoptosis, promotion of pancreatic β cell proliferation, and stimulation of insulin secretion. Further screening of bone-derived factors revealed that islet regeneration-derived protein III gamma (RegIIIγ) is an osteoblast-derived hypoxia-sensing factor critical for protection against STZ-induced T1DM. In addition, we found that iminodiacetic acid deferoxamine (SF-DFO), a compound that mimics hypoxia and targets bone tissue, can alleviate symptoms of STZ-induced T1DM by activating the HIF-1α-RegIIIγ pathway in the skeleton. These data suggest that the osteoblastic HIF-1α-RegIIIγ pathway is a potential target for treating T1DM.
Collapse
Affiliation(s)
- Minglong Qiu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Leilei Chang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Guoqing Tang
- Kunshan Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, 388 Zuchongzhi Road, Kunshan, 215300, Jiangsu, China
| | - Wenkai Ye
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yiming Xu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Nijiati Tulufu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Zhou Dan
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jin Qi
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Lianfu Deng
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Changwei Li
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
15
|
Hiasa M, Endo I, Matsumoto T. Bone-fat linkage via interleukin-11 in response to mechanical loading. J Bone Miner Metab 2024; 42:447-454. [PMID: 38324177 DOI: 10.1007/s00774-023-01493-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/10/2023] [Indexed: 02/08/2024]
Abstract
Positive regulators of bone formation, such as mechanical loading and PTH, stimulate and negative regulators, such as aging and glucocorticoid excess, suppress IL-11 gene transcription in osteoblastic cells. Signal transduction from mechanical loading and PTH stimulation involves two pathways: one is Ca2+-ERK-CREB pathway which facilitates binding of ∆FosB/JunD to the AP-1 site to enhance IL-11 gene transcription, and the other is Smad1/5 phosphorylation that promotes IL-11 gene transcription via SBE binding and complex formation with ∆FosB/JunD. The increased IL-11 suppresses Sost expression via IL-11Rα-STAT1/3-HDAC4/5 pathway and enhances Wnt signaling in the bone to stimulate bone formation. Thus, IL-11 mediates stimulatory and inhibitory signals of bone formation by affecting Wnt signaling. Physiologically important stimulation of bone formation is exercise-induced mechanical loading, but exercise simultaneously requires energy source for muscle contraction. Exercise-induced stimulation of IL-11 expression in the bone increases the secretion of IL-11 from the bone. The increased circulating IL-11 acts like a hormone to enhance adipolysis as an energy source with a reduction in adipogenic differentiation via a suppression of Dkk1/2 expression in the adipose tissue. Such bone-fat linkage can be a mechanism whereby exercise increases bone mass and, at the same time, maintains energy supply from the adipose tissue.
Collapse
Affiliation(s)
- Masahiro Hiasa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Dentistry, Tokushima, 770-8503, Japan
| | - Itsuro Endo
- Department of Endocrinology, Metabolism and Hematology, Tokushima University Graduate School of Medical Sciences, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503,, Japan
| | - Toshio Matsumoto
- Department of Endocrinology, Metabolism and Hematology, Tokushima University Graduate School of Medical Sciences, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503,, Japan.
| |
Collapse
|
16
|
Chen J, Hao Z, Li H, Wang J, Chen T, Wang Y, Shi G, Wang J, Wang Z, Zhang Z, Li J. Osteoporotic osseointegration: therapeutic hallmarks and engineering strategies. Theranostics 2024; 14:3859-3899. [PMID: 38994021 PMCID: PMC11234277 DOI: 10.7150/thno.96516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024] Open
Abstract
Osteoporosis is a systemic skeletal disease caused by an imbalance between bone resorption and formation. Current treatments primarily involve systemic medication and hormone therapy. However, these systemic treatments lack directionality and are often ineffective for locally severe osteoporosis, with the potential for complex adverse reactions. Consequently, treatment strategies using bioactive materials or external interventions have emerged as the most promising approaches. This review proposes twelve microenvironmental treatment targets for osteoporosis-related pathological changes, including local accumulation of inflammatory factors and reactive oxygen species (ROS), imbalance of mitochondrial dynamics, insulin resistance, disruption of bone cell autophagy, imbalance of bone cell apoptosis, changes in neural secretions, aging of bone cells, increased local bone tissue vascular destruction, and decreased regeneration. Additionally, this review examines the current research status of effective or potential biophysical and biochemical stimuli based on these microenvironmental treatment targets and summarizes the advantages and optimal parameters of different bioengineering stimuli to support preclinical and clinical research on osteoporosis treatment and bone regeneration. Finally, the review addresses ongoing challenges and future research prospects.
Collapse
Affiliation(s)
- Jiayao Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Jianping Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | - Guang Shi
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zepu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zheyuan Zhang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| |
Collapse
|
17
|
Li SY, Xue ST, Li ZR. Osteoporosis: Emerging targets on the classical signaling pathways of bone formation. Eur J Pharmacol 2024; 973:176574. [PMID: 38642670 DOI: 10.1016/j.ejphar.2024.176574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/30/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
Osteoporosis is a multifaceted skeletal disorder characterized by reduced bone mass and structural deterioration, posing a significant public health challenge, particularly in the elderly population. Treatment strategies for osteoporosis primarily focus on inhibiting bone resorption and promoting bone formation. However, the effectiveness and limitations of current therapeutic approaches underscore the need for innovative methods. This review explores emerging molecular targets within crucial signaling pathways, including wingless/integrated (WNT), bone morphogenetic protein (BMP), hedgehog (HH), and Notch signaling pathway, to understand their roles in osteogenesis regulation. The identification of crosstalk targets between these pathways further enhances our comprehension of the intricate bone metabolism cycle. In summary, unraveling the molecular complexity of osteoporosis provides insights into potential therapeutic targets beyond conventional methods, offering a promising avenue for the development of new anabolic drugs.
Collapse
Affiliation(s)
- Si-Yan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Si-Tu Xue
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Zhuo-Rong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
18
|
Yadalam PK, Ramadoss R, Suresh R. Weighted Gene Co-expression Network Analysis of the Inflammatory Wnt Signaling Reveals Biomarkers Related to Bone Formation. Cureus 2024; 16:e63510. [PMID: 39081453 PMCID: PMC11288288 DOI: 10.7759/cureus.63510] [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: 05/15/2024] [Accepted: 06/29/2024] [Indexed: 08/02/2024] Open
Abstract
Background and aim Osteocytes regulate bone metabolism and balance through various mechanisms, including the Wnt (Wingless-related integration site signal transduction) signaling pathway. Weighted gene co-expression network analysis (WGCNA) is a computational method to identify functionally related genes based on expression patterns, especially in the Wnt-beta-catenin and osteo-regenerative pathways. This study aims to analyze gene modules of the Wnt signaling pathway from WGCNA analysis. Methods The study used a microarray dataset from the GEO (GSE228306) to analyze differential gene expression in human primary monocytes. The study standardized datasets using Robust Multi-Array Average (RMA) expression measure and Integrated Differential Expression and Pathway (IDEP) analysis tool, building a co-expression network for group-specific component (GC) genes. Results The study uses WGCNA to identify co-expression modules with dysregulated mRNAs, revealing enrichment in Wnt-associated pathways and top hub-enriched genes like colony-stimulating factor 3 (CSF3), interleukin-6 (IL-6), IL-23 subunit alpha (IL23A), suppressor of cytokine signaling 1 (SOCS1), and C-C motif chemokine ligand 19 (CCL19). Conclusion WGCNA analysis of the Wnt signaling pathway will involve functional annotation, network visualization, validation, integration with other omics data, and addressing method limitations for better understanding.
Collapse
Affiliation(s)
- Pradeep Kumar Yadalam
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Ramya Ramadoss
- Oral Pathology and Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Ramya Suresh
- Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
19
|
Kanai M, Ganbaatar B, Endo I, Ohnishi Y, Teramachi J, Tenshin H, Higa Y, Hiasa M, Mitsui Y, Hara T, Masuda S, Yamagami H, Yamaguchi Y, Aihara KI, Sebe M, Tsutsumi R, Sakaue H, Matsumoto T, Abe M. Inflammatory Cytokine-Induced Muscle Atrophy and Weakness Can Be Ameliorated by an Inhibition of TGF-β-Activated Kinase-1. Int J Mol Sci 2024; 25:5715. [PMID: 38891908 PMCID: PMC11172090 DOI: 10.3390/ijms25115715] [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: 04/28/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic inflammation causes muscle wasting. Because most inflammatory cytokine signals are mediated via TGF-β-activated kinase-1 (TAK1) activation, inflammatory cytokine-induced muscle wasting may be ameliorated by the inhibition of TAK1 activity. The present study was undertaken to clarify whether TAK1 inhibition can ameliorate inflammation-induced muscle wasting. SKG/Jcl mice as an autoimmune arthritis animal model were treated with a small amount of mannan as an adjuvant to enhance the production of TNF-α and IL-1β. The increase in these inflammatory cytokines caused a reduction in muscle mass and strength along with an induction of arthritis in SKG/Jcl mice. Those changes in muscle fibers were mediated via the phosphorylation of TAK1, which activated the downstream signaling cascade via NF-κB, p38 MAPK, and ERK pathways, resulting in an increase in myostatin expression. Myostatin then reduced the expression of muscle proteins not only via a reduction in MyoD1 expression but also via an enhancement of Atrogin-1 and Murf1 expression. TAK1 inhibitor, LL-Z1640-2, prevented all the cytokine-induced changes in muscle wasting. Thus, TAK1 inhibition can be a new therapeutic target of not only joint destruction but also muscle wasting induced by inflammatory cytokines.
Collapse
Affiliation(s)
- Mai Kanai
- Department of Bioregulatory Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan;
| | - Byambasuren Ganbaatar
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| | - Itsuro Endo
- Department of Bioregulatory Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan;
| | - Yukiyo Ohnishi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| | - Jumpei Teramachi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
- Department of Oral Function and Anatomy, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8570, Japan
| | - Hirofumi Tenshin
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (H.T.); (Y.H.); (M.H.)
| | - Yoshiki Higa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (H.T.); (Y.H.); (M.H.)
| | - Masahiro Hiasa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (H.T.); (Y.H.); (M.H.)
| | - Yukari Mitsui
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| | - Tomoyo Hara
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| | - Shiho Masuda
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| | - Hiroki Yamagami
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| | - Yuki Yamaguchi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| | - Ken-ichi Aihara
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan;
| | - Mayu Sebe
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Okayama 700-8570, Japan;
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (R.T.); (H.S.)
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (R.T.); (H.S.)
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima 770-8503, Japan;
| | - Masahiro Abe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; (B.G.); (Y.O.); (J.T.); (Y.M.); (T.H.); (S.M.); (H.Y.); (Y.Y.); (M.A.)
| |
Collapse
|
20
|
Shen Y, Wang H, Xie H, Zhang J, Ma Q, Wang S, Yuan P, Xue H, Hong H, Fan S, Xu W, Xie Z. l-arginine promotes angio-osteogenesis to enhance oxidative stress-inhibited bone formation by ameliorating mitophagy. J Orthop Translat 2024; 46:53-64. [PMID: 38808262 PMCID: PMC11131000 DOI: 10.1016/j.jot.2024.03.003] [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: 10/26/2023] [Revised: 02/03/2024] [Accepted: 03/04/2024] [Indexed: 05/30/2024] Open
Abstract
Background Osteoporosis is one of the most common bone diseases in middle-aged and elderly populations worldwide. The development of new drugs to treat the disease is a key focus of research. Current treatments for osteoporosis are mainly directed at promoting osteoblasts and inhibiting osteoclasts. However, there is currently no ideal approach for osteoporosis treatment. l-arginine is a semi-essential amino acid involved in a number of cellular processes, including nitric production, protein biosynthesis, and immune responses. We previously reported that l-arginine-derived compounds can play a regulatory role in bone homeostasis. Purpose To investigate the specific effect of l-arginine on bone homeostasis. Methods Mildly aged and ovariectomized mouse models were used to study the effects of l-arginine on osteogenesis and angiogenesis, assessed by micro-computed tomography and immunostaining of bone tissue. The effect of l-arginine on osteogenesis, angiogenesis, and adipogenesis was further studied in vitro using osteoblasts obtained from cranial cap bone, endothelial cells, and an adipogenic cell line. Specific methods to assess these processes included lipid staining, cell migration, tube-forming, and wound-healing assays. Protein and mRNA expression was determined for select biomarkers. Results We found that l-arginine attenuated bone loss and promoted osteogenesis and angiogenesis. l-arginine increased the activity of vascular endothelial cells, whereas it inhibited adipogenesis in vitro. In addition, we found that l-arginine altered the expression of PINK1/Parkin and Bnip3 in the mitochondria of osteoblast-lineage and endothelial cells, thereby promoting mitophagy and protecting cells from ROS. Similarly, l-arginine treatment effectively ameliorated osteoporosis in an ovariectomized mouse model. Conclusion l-arginine promotes angio-osteogenesis, and inhibits adipogenesis, effects mediated by the PINK1/Parkin- and Bnip3-mediated mitophagy. The Translational Potential of this Article L-arginine supplementation may be an effective adjunct therapy in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Yang Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Haoming Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hongwei Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jiateng Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Qingliang Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shiyu Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hong Xue
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Huaxing Hong
- Department of Orthopaedics, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Taizhou, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Wenbin Xu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
21
|
Ma H, Cai X, Hu J, Song S, Zhu Q, Zhang Y, Ma R, Shen D, Yang W, Zhou P, Zhang D, Luo Q, Hong J, Li N. Association of systemic inflammatory response index with bone mineral density, osteoporosis, and future fracture risk in elderly hypertensive patients. Postgrad Med 2024; 136:406-416. [PMID: 38753519 DOI: 10.1080/00325481.2024.2354158] [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: 04/01/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVES This study sought to investigate the relationship between the systemic inflammatory response index (SIRI) and bone mineral density (BMD), osteoporosis, and future fracture risk in elderly hypertensive patients. METHODS Elderly hypertensive patients (age ≥60 years) who attended our hospital between January 2021 and December 2023 and completed BMD screening were included in the study. Analyses were performed with multivariate logistic and linear regression. RESULTS The multiple linear regression indicated that SIRI levels were significantly negatively correlated with lumbar 1 BMD (β = -0.15, 95% CI: -0.24, -0.05), lumbar 2 BMD (β = -0.15, 95% CI: -0.24, -0.05), lumbar 3 BMD (β = -1.35, 95% CI: -0.23, -0.02), lumbar 4 BMD (β = -0.11, 95% CI: -0.30, -0.10), femur neck BMD (β = -0.11, 95% CI: -0.18, -0.05) and Ward's triangle BMD (β = -0.12, 95% CI: -0.20, -0.05) among elderly hypertensive patients, after fully adjusting for confounders. Furthermore, we observed that SIRI was positively associated with future fracture risk in elderly hypertensive patients. Specifically, SIRI was associated with an increased risk of major osteoporotic fractures (β = 0.33) and hip fractures (β = 0.25). The logistic regression analysis indicated that there is an association between the SIRI level and an increased risk of osteoporosis (OR = 1.60, 95% CI = 1.37, 1.87), after fully adjusting for confounders. CONCLUSIONS Our findings indicate a potential association between SIRI and BMD, osteoporosis, and the risk of future fractures in elderly hypertensive patients. However, further studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Huimin Ma
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Xintian Cai
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Junli Hu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Shuaiwei Song
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Qing Zhu
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Yingying Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Rui Ma
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Di Shen
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Wenbo Yang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Pan Zhou
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Delian Zhang
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Qin Luo
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Jing Hong
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Nanfang Li
- Hypertension Center of People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, NHC Key Laboratory of Hypertension Clinical Research, Key Laboratory of Xinjiang Uygur Autonomous Region "Hypertension Research Laboratory", Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| |
Collapse
|
22
|
Sims NA, Griffin MDW. Craniosynostosis-associated variants in the IL-11R complex: new insights and questions. FEBS J 2024; 291:1663-1666. [PMID: 38329021 DOI: 10.1111/febs.17078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
Skull growth involves the expansion of both the flat calvarial bones of the skull and the fibrous marginal zones, termed sutures, between them. This process depends on co-ordinated proliferation of mesenchymal-derived progenitor cells within the sutures, and their differentiation to osteoblasts which produce the bone matrix required to expand the size of the bony plates. Defects lead to premature closure of these sutures, termed craniosynostosis, resulting in heterogeneous head shape differences due to restricted growth of one or more sutures. The impact on the individual depends on how many and which sutures are affected and the severity of the effect. Several genetic loci are responsible, including a wide range of variants in the gene for the interleukin 11 receptor (IL11RA, OMIM#600939). Recent work from Kespohl and colleagues provides new insights into how some of these variants influence IL-11R function; we discuss their influences on IL-11R structure and IL-11 function as a stimulus of osteoblast differentiation.
Collapse
Affiliation(s)
- Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Australia
- Mary Mackillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Michael D W Griffin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Australia
| |
Collapse
|
23
|
Du B, Luo S, Zhu X, Hu M, Feng X, Yu Q, Bai B, Xu J, Wang J. WAY-262611 ameliorates the inflammatory bowel disease by activating Wnt/β-catenin pathway. In Vitro Cell Dev Biol Anim 2024; 60:128-138. [PMID: 38393664 DOI: 10.1007/s11626-023-00809-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/21/2023] [Indexed: 02/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a non-specific and relapsing intestinal inflammation. The injury and repair of intestinal epithelial together determine the occurrence and development of IBD. Wnt/β-catenin pathway is considered as the key role in the proliferation and differentiation of intestinal stem cells which is negative regulated by Dickkiop (DKKs). WAY-262611 is a novel inhibitor of DKK-1, and has demonstrated therapeutic effect on some disease including osteoporosis. Thus, we investigated the effect of WAY-262611 on IBD. Firstly, a mice model of IBD was established by DSS induction, by which the expression of Wnt3a and DKK-1 were detected by immumohistochemical staining to display their correlation. Next, using WAY-262611 the ameliorative effect on IBD was validated by histopathological staining. Using Mode-k cells the experiments in vitro were also conducted, in which the viability and apoptosis were determined. By detecting expression of Wnt3a and DKK-1 and observing nuclear translocation of β-catenin, the activation of Wnt/β-catenin pathway was validated. Finally, the incidence of the orthotopic colorectal cancer was calculated under continuous administration by DSS. Results demonstrated that the expression of Wnt3a is negative correlated with DKK-1. WAY-262611 ameliorated the IBD and reduced apoptosis of Mode-k cells induced by DSS. The protective effect of WAY-262611 on Mode-k cells is mediated by Wnt/β-catenin pathway activation. In addition, WAY-262611 lowered the incidence rate of orthotopic colorectal cancer. All these results concluded that WAY-262611 could mitigate the IBD by activating Wnt/β-catenin pathway in mice.
Collapse
Affiliation(s)
- Baiyinzi Du
- Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
- Tianjin General Hospital, Tianjin Medical University, Tianjin, China
| | - Shudan Luo
- College of Basic Medicine, Naval Medical University, Xiangyin Road, 200433, Shanghai, People's Republic of China
| | - Xujun Zhu
- Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Maqing Hu
- College of Basic Medicine, Naval Medical University, Xiangyin Road, 200433, Shanghai, People's Republic of China
| | - Xianzhang Feng
- College of Basic Medicine, Naval Medical University, Xiangyin Road, 200433, Shanghai, People's Republic of China
| | - Qianjun Yu
- College of Basic Medicine, Naval Medical University, Xiangyin Road, 200433, Shanghai, People's Republic of China
| | - Bin Bai
- Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China.
| | - Jian Xu
- Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Jun Wang
- Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| |
Collapse
|
24
|
Xu K, Fei W, Gao W, Fan C, Li Y, Hong Y, Cui R. SOD3 regulates FLT1 to affect bone metabolism by promoting osteogenesis and inhibiting adipogenesis through PI3K/AKT and MAPK pathways. Free Radic Biol Med 2024; 212:65-79. [PMID: 38141889 DOI: 10.1016/j.freeradbiomed.2023.12.021] [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: 07/09/2023] [Revised: 11/30/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023]
Abstract
Osteoporosis is a chronic disease that seriously affects the quality of life and longevity of the elderly, so exploring the mechanism of osteoporosis is crucial for drug development and treatment. Bone marrow mesenchymal stem cells are stem cells with multiple differentiation potentials in bone marrow, and changing their differentiation direction can change bone mass. As an extracellular superoxide dismutase, Superoxide Dismutase 3 (SOD3) has been proved to play an important role in multiple organs, but the detailed mechanism of action in bone metabolism is still unclear. In this study, the results of clinical serum samples ELISA and single cell sequencing chip analysis proved that the expression of SOD3 was positively correlated with bone mass, and SOD3 was mainly expressed in osteoblasts and adipocytes and rarely expressed in osteoblasts in BMSCs. In vitro experiments showed that SOD3 can promote osteogenesis and inhibit adipogenesis. Compared with WT mice, the mice that were knocked out of SOD3 had a significant decrease in bone mineral density and significant changes in related parameters. The results of HE and IHC staining suggested that knocking out SOD3 would lead to fat accumulation in the bone marrow cavity and weakened osteogenesis. Both in vitro and in vivo experiments indicated that SOD3 affects bone metabolism by promoting osteogenesis and inhibiting adipogenesis. The results of transcriptome sequencing and revalidation showed that SOD3 can affect the expression of FLT1. Through in vitro experiments, we proved that FLT1 can also promote osteogenesis and inhibit adipogenesis. In addition, through the repeated experiments, the interaction between the two molecules (SOD3 and FLT1) was verified again. Finally, it was verified by WB that SOD3 regulates FLT1 to affect bone metabolism through PI3K/AKT and MAPK pathways.
Collapse
Affiliation(s)
- Ke Xu
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Aging and Medicine, Shanghai, China; Center of Community-Based Health Research, Fudan University, Shanghai, China.
| | - Wenchao Fei
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Aging and Medicine, Shanghai, China; Center of Community-Based Health Research, Fudan University, Shanghai, China.
| | - Wenxue Gao
- Medical Services Section, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Changxiu Fan
- Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China.
| | - Yinghua Li
- Shanghai Clinical Research Center for Aging and Medicine, Shanghai, China; Center of Community-Based Health Research, Fudan University, Shanghai, China; Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China.
| | - Yang Hong
- Department of Orthopedics, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China; Shanghai Clinical Research Center for Aging and Medicine, Shanghai, China; Center of Community-Based Health Research, Fudan University, Shanghai, China.
| | - Ran Cui
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Han Y, Gao H, Gan X, Liu J, Bao C, He C. Roles of IL-11 in the regulation of bone metabolism. Front Endocrinol (Lausanne) 2024; 14:1290130. [PMID: 38352248 PMCID: PMC10862480 DOI: 10.3389/fendo.2023.1290130] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/29/2023] [Indexed: 02/16/2024] Open
Abstract
Bone metabolism is the basis for maintaining the normal physiological state of bone, and imbalance of bone metabolism can lead to a series of metabolic bone diseases. As a member of the IL-6 family, IL-11 acts primarily through the classical signaling pathway IL-11/Receptors, IL-11 (IL-11R)/Glycoprotein 130 (gp130). The regulatory role of IL-11 in bone metabolism has been found earlier, but mainly focuses on the effects on osteogenesis and osteoclasis. In recent years, more studies have focused on IL-11's roles and related mechanisms in different bone metabolism activities. IL-11 regulates osteoblasts, osteoclasts, BM stromal cells, adipose tissue-derived mesenchymal stem cells, and chondrocytes. It's involved in bone homeostasis, including osteogenesis, osteolysis, bone marrow (BM) hematopoiesis, BM adipogenesis, and bone metastasis. This review exams IL-11's role in pathology and bone tissue, the cytokines and pathways that regulate IL-11 expression, and the feedback regulations of these pathways.
Collapse
Affiliation(s)
| | | | - Xinling Gan
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | | | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Zhang Y, Chen Q. Novel insights into osteocyte and inter-organ/tissue crosstalk. Front Endocrinol (Lausanne) 2024; 14:1308408. [PMID: 38685911 PMCID: PMC11057460 DOI: 10.3389/fendo.2023.1308408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/14/2023] [Indexed: 05/02/2024] Open
Abstract
Osteocyte, a cell type living within the mineralized bone matrix and connected to each other by means of numerous dendrites, appears to play a major role in body homeostasis. Benefiting from the maturation of osteocyte extraction and culture technique, many cross-sectional studies have been conducted as a subject of intense research in recent years, illustrating the osteocyte-organ/tissue communication not only mechanically but also biochemically. The present review comprehensively evaluates the new research work on the possible crosstalk between osteocyte and closely situated or remote vital organs/tissues. We aim to bring together recent key advances and discuss the mutual effect of osteocyte and brain, kidney, vascular calcification, muscle, liver, adipose tissue, and tumor metastasis and elucidate the therapeutic potential of osteocyte.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingchang Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Research Center for Medical Imaging in Hubei Province, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
27
|
Zhao F, Zhang Y, Pei S, Wang S, Hu L, Wang L, Qian A, Yang TL, Guo Y. Mechanobiological crosstalk among bone cells and between bone and other organs. BONE CELL BIOMECHANICS, MECHANOBIOLOGY AND BONE DISEASES 2024:215-247. [DOI: 10.1016/b978-0-323-96123-3.00015-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
28
|
Gugala Z. What's New in Musculoskeletal Basic Science. J Bone Joint Surg Am 2023; 105:1831-1836. [PMID: 38063778 DOI: 10.2106/jbjs.23.01003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Affiliation(s)
- Zbigniew Gugala
- Department of Orthopaedic Surgery and Rehabilitation, The University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
29
|
Dong B, Zhu J, Chen X, Jiang H, Deng Y, Xu L, Wang Y, Li S. The Emerging Role of Interleukin-(IL)-11/IL-11R in Bone Metabolism and Homeostasis: From Cytokine to Osteokine. Aging Dis 2023; 14:2113-2126. [PMID: 37199584 PMCID: PMC10676798 DOI: 10.14336/ad.2023.0306] [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: 01/03/2023] [Accepted: 03/06/2023] [Indexed: 05/19/2023] Open
Abstract
Interleukin-(IL)-11 is a cytokine involved in hematopoiesis, cancer metastasis, and inflammation. IL-11 belongs to the IL-6 cytokine family, binding to the complex of receptors glycoprotein gp130 and the ligand-specific-receptor subunits (IL-11Rα or their soluble counterpart sIL-11R). IL-11/IL-11R signaling enhances osteoblast differentiation and bone formation and mitigates osteoclast-induced bone resorption and cancer bone metastasis. Recent studies have shown that systemic and osteoblast/osteocyte-specific IL-11 deficiency leads to reduced bone mass and formation, but also adiposity, glucose intolerance, and insulin resistance. In humans, mutations of IL-11 and the receptor IL-11RA genes are associated with height reduction, osteoarthritis, and craniosynostosis. In this review, we describe the emerging role of IL-11/IL-11R signaling in bone metabolism by targeting osteoblasts, osteoclasts, osteocytes, and bone mineralization. Furthermore, IL-11 promotes osteogenesis and suppresses adipogenesis, thereby influencing the fate of osteoblast/adipocyte differentiation derived from pluripotent mesenchymal stem cells. We have newly identified IL-11 as a bone-derived cytokine that regulates bone metabolism and the link between bone and other organs. Thus, IL-11 is vital in bone homeostasis and could be considered a potential therapeutic strategy.
Collapse
Affiliation(s)
- Bingzi Dong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingjing Zhu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xian Chen
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongyuan Jiang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yujie Deng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shufa Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
30
|
Torres HM, Arnold KM, Oviedo M, Westendorf JJ, Weaver SR. Inflammatory Processes Affecting Bone Health and Repair. Curr Osteoporos Rep 2023; 21:842-853. [PMID: 37759135 PMCID: PMC10842967 DOI: 10.1007/s11914-023-00824-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing. RECENT FINDINGS Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells. Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.
Collapse
Affiliation(s)
- Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Biomedical Engineering and Physiology Track/Regenerative Sciences Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuela Oviedo
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
31
|
Panahipour L, Abbasabadi AO, Wagner A, Kratochwill K, Pichler M, Gruber R. Bone Allograft Acid Lysates Change the Genetic Signature of Gingival Fibroblasts. Int J Mol Sci 2023; 24:16181. [PMID: 38003371 PMCID: PMC10671348 DOI: 10.3390/ijms242216181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Bone allografts are widely used as osteoconductive support to guide bone regrowth. Bone allografts are more than a scaffold for the immigrating cells as they maintain some bioactivity of the original bone matrix. Yet, it remains unclear how immigrating cells respond to bone allografts. To this end, we have evaluated the response of mesenchymal cells exposed to acid lysates of bone allografts (ALBA). RNAseq revealed that ALBA has a strong impact on the genetic signature of gingival fibroblasts, indicated by the increased expression of IL11, AREG, C11orf96, STC1, and GK-as confirmed by RT-PCR, and for IL11 and STC1 by immunoassays. Considering that transforming growth factor-β (TGF-β) is stored in the bone matrix and may have caused the expression changes, we performed a proteomics analysis, TGF-β immunoassay, and smad2/3 nuclear translocation. ALBA neither showed detectable TGF-β nor was the lysate able to induce smad2/3 translocation. Nevertheless, the TGF-β receptor type I kinase inhibitor SB431542 significantly decreased the expression of IL11, AREG, and C11orf96, suggesting that other agonists than TGF-β are responsible for the robust cell response. The findings suggest that IL11, AREG, and C11orf96 expression in mesenchymal cells can serve as a bioassay reflecting the bioactivity of the bone allografts.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Anja Wagner
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus Kratochwill
- Core Facility Proteomics, Medical University of Vienna, 1090 Vienna, Austria; (A.W.); (K.K.)
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
32
|
Panahipour L, Abbasabadi AO, Gruber R. Gingival Fibroblasts Are Sensitive to Oral Cell Lysates Indicated by Their IL11 Expression. Bioengineering (Basel) 2023; 10:1193. [PMID: 37892923 PMCID: PMC10604186 DOI: 10.3390/bioengineering10101193] [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: 08/25/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Damaged cells that appear as a consequence of invasive dental procedures or in response to dental materials are supposed to release damage-associated signals. These damage-associated signals not only support tissue regeneration but might also contribute to unwanted fibrosis. The aim of this study was to identify a molecular target that reflects how fibroblasts respond to necrotic oral tissue cells. To simulate the cell damage, we prepared necrotic cell lysates by sonication of the osteocytic cell line IDG-SW3 and exposed them to gingival fibroblasts. RNAseq revealed a moderate increase in IL11 expression in the gingival fibroblasts, a pleiotropic cytokine involved in fibrosis and inflammation, and also in regeneration following trauma. Necrotic lysates of the human squamous carcinoma cell lines HSC2 and TR146, as well as of gingival fibroblasts, however, caused a robust increase in IL11 expression in the gingival fibroblasts. Consistently, immunoassay revealed significantly increased IL11 levels in the gingival fibroblasts when exposed to the respective lysates. Considering that IL11 is a TGF-β target gene, IL11 expression was partially blocked by SB431542, a TGF-β receptor type I kinase inhibitor. Moreover, lysates from the HSC2, TR146, and gingival fibroblasts caused a moderate smad2/3 nuclear translocation in the gingival fibroblasts. Taken together and based on IL11 expression, our findings show that fibroblasts are sensitive to damaged oral tissue cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (A.O.A.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
33
|
Laget J, Cortijo I, Boukhaled JH, Muyor K, Duranton F, Jover B, Raynaud F, Lajoix AD, Argilés À, Gayrard N. Cafeteria Diet-Induced Obesity Worsens Experimental CKD. Nutrients 2023; 15:3331. [PMID: 37571269 PMCID: PMC10421241 DOI: 10.3390/nu15153331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Obesity is a significant risk factor for chronic kidney disease (CKD). This study aimed to evaluate the impact of obesity on the development of kidney fibrosis in a model of cafeteria diet rats undergoing 5/6th nephrectomy (SNx). Collagen 1, 3, and 4 expression, adipocyte size, macrophage number, and the expression of 30 adipokines were determined. Collagen 1 expression in kidney tissue was increased in Standard-SNx and Cafeteria-SNx (7.1 ± 0.6% and 8.9 ± 0.9 tissue area, respectively). Renal expression of collagen 3 and 4 was significantly increased (p < 0.05) in Cafeteria-SNx (8.6 ± 1.5 and 10.9 ± 1.9% tissue area, respectively) compared to Cafeteria (5.2 ± 0.5 and 6.3 ± 0.6% tissue area, respectively). Adipocyte size in eWAT was significantly increased by the cafeteria diet. In Cafeteria-SNx, we observed a significant increase in macrophage number in the kidney (p = 0.01) and a consistent tendency in eWAT. The adipokine level was higher in the Cafeteria groups. Interleukin 11, dipeptidyl peptidase 4, and serpin 1 were increased in Cafeteria-SNx. In the kidney, collagen 3 and 4 expressions and the number of macrophages were increased in Cafeteria-SNx, suggesting an exacerbation by preexisting obesity of CKD-induced renal inflammation and fibrosis. IL11, DPP4, and serpin 1 can act directly on fibrosis and participate in the observed worsening CKD.
Collapse
Affiliation(s)
- Jonas Laget
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| | - Irene Cortijo
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| | - Juliana H. Boukhaled
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| | - Karen Muyor
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| | - Flore Duranton
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| | - Bernard Jover
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| | - Fabrice Raynaud
- PhyMedExp, INSERM, CNRS, Université de Montpellier, 34090 Montpellier, France;
| | - Anne-Dominique Lajoix
- Biocommunication in Cardio-Metabolism (BC2M), University of Montpellier, 34090 Montpellier, France;
| | - Àngel Argilés
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| | - Nathalie Gayrard
- RD-Néphrologie, 34090 Montpellier, France; (J.L.); (I.C.); (J.H.B.); (K.M.); (F.D.); (B.J.); (À.A.)
| |
Collapse
|
34
|
Xu J, Yu L, Liu F, Wan L, Deng Z. The effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis: a review. Front Immunol 2023; 14:1222129. [PMID: 37475866 PMCID: PMC10355373 DOI: 10.3389/fimmu.2023.1222129] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/12/2023] [Indexed: 07/22/2023] Open
Abstract
The complicated connections and cross talk between the skeletal system and the immune system are attracting more attention, which is developing into the field of Osteoimmunology. In this field, cytokines that are among osteoblasts and osteoclasts play a critical role in bone remodeling, which is a pathological process in the pathogenesis and development of osteoporosis. Those cytokines include the tumor necrosis factor (TNF) family, the interleukin (IL) family, interferon (IFN), chemokines, and so on, most of which influence the bone microenvironment, osteoblasts, and osteoclasts. This review summarizes the effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis, aiming to providing the latest reference to the role of immunology in osteoporosis.
Collapse
Affiliation(s)
- Jie Xu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Linxin Yu
- Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Longbiao Wan
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Deng
- Hubei Provincial Hospital of Traditional Chinese Medicine (TCM), Wuhan, China
| |
Collapse
|
35
|
Kitase Y, Prideaux M. Regulation of the Osteocyte Secretome with Aging and Disease. Calcif Tissue Int 2023; 113:48-67. [PMID: 37148298 DOI: 10.1007/s00223-023-01089-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
As the most numerous and long-lived of all bone cells, osteocytes have essential functions in regulating skeletal health. Through the lacunar-canalicular system, secreted proteins from osteocytes can reach cells throughout the bone. Furthermore, the intimate connectivity between the lacunar-canalicular system and the bone vasculature allows for the transport of osteocyte-secreted factors into the circulation to reach the entire body. Local and endocrine osteocyte signaling regulates physiological processes such as bone remodeling, bone mechanoadaptation, and mineral homeostasis. However, these processes are disrupted by impaired osteocyte function induced by aging and disease. Dysfunctional osteocyte signaling is now associated with the pathogenesis of many disorders, including chronic kidney disease, cancer, diabetes mellitus, and periodontitis. In this review, we focus on the targeting of bone and extraskeletal tissues by the osteocyte secretome. In particular, we highlight the secreted osteocyte proteins, which are known to be dysregulated during aging and disease, and their roles during disease progression. We also discuss how therapeutic or genetic targeting of osteocyte-secreted proteins can improve both skeletal and systemic health.
Collapse
Affiliation(s)
- Yukiko Kitase
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
36
|
Zhang Z, Zhang Z, Shu H, Meng Y, Lin T, Ma J, Zhao J, Zhou X. Association between gut microbiota and bone metabolism: Insights from bibliometric analysis. Front Physiol 2023; 14:1156279. [PMID: 37153210 PMCID: PMC10154530 DOI: 10.3389/fphys.2023.1156279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
Gut microbiota has been reported to participate in bone metabolism. However, no article has quantitatively and qualitatively analyzed this crossing field. The present study aims to analyze the current international research trends and demonstrate possible hotspots in the recent decade through bibliometrics. We screened out 938 articles meeting the standards from 2001 to 2021 in the Web of Science Core Collection database. Bibliometric analyses were performed and visualized using Excel, Citespace, and VOSviewer. Generally, the annual number of published literatures in this field shows an escalating trend. The United States has the largest number of publications, accounting for 30.4% of the total. Michigan State University and Sichuan University have the largest number of publications, while Michigan State University has the highest average number of citations at 60.00. Nutrients published 49 articles, ranking first, while the Journal of Bone and Mineral Research had the highest average number of citations at 13.36. Narayanan Parameswaran from Michigan State University, Roberto Pacifici from Emory University, and Christopher Hernandez from Cornell University were the three professors who made the largest contribution to this field. Frequency analysis showed that inflammation (148), obesity (86), and probiotics (81) are keywords with the highest focus. Moreover, keywords cluster analysis and keywords burst analysis showed that "inflammation", "obesity", and "probiotics" were the most researched topics in the field of gut microbiota and bone metabolism. Scientific publications related to gut microbiota and bone metabolism have continuously risen from 2001 to 2021. The underlying mechanism has been widely studied in the past few years, and factors affecting the alterations of the gut microbiota, as well as probiotic treatment, are emerging as new research trends.
Collapse
Affiliation(s)
- Zhanrong Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Orthopedic Rehabilitation, Qingdao Special Servicemen Recuperation Center of PLA Navy, Qingdao, China
| | - Haoming Shu
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yichen Meng
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Tao Lin
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Jun Ma
- Department of Orthopedics, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jun Ma, ; Jianquan Zhao, ; Xuhui Zhou,
| | - Jianquan Zhao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Jun Ma, ; Jianquan Zhao, ; Xuhui Zhou,
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- Translational Research Center of Orthopedics, Shanghai General Hospital (Shanghai First People’s Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jun Ma, ; Jianquan Zhao, ; Xuhui Zhou,
| |
Collapse
|