1
|
Huang Z, Zhang L, Luo M, Zhang X, Huang Y, Tian Y, Wu Z, Li X. Whole-genome resequencing reveals genetic differentiation and selection signatures among wild, local and commercial duck populations. Anim Biosci 2025; 38:898-909. [PMID: 40336248 PMCID: PMC12062807 DOI: 10.5713/ab.24.0643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/20/2024] [Accepted: 11/27/2024] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE The purpose of this study was to systematically analyze the population genetic structure and genetic diversity among wild, local and commercial populations using whole-genome sequencing data from 416 individuals of 22 duck breeds in China and to further explore genetic pathways and candidate genes associated with importantly economic traits. METHODS We performed principal component analysis, an unrooted neighbor-joining phylogenetic tree and ADMIXTURE to analyze the population structure. We compared the genetic diversity among wild, local and commercial populations using the effective population size, inbreeding coefficient, expected heterozygosity, observed heterozygosity, nucleotide diversity and regions of homozygosity. To detect selection signatures, we calculated the locus-specific branch length for local and commercial populations and calculated genetic differentiation coefficient and genetic diversity between egg and dual-purpose breeds. RESULTS Wild, local and commercial duck populations formed three distinct genetic groups. The commercial population presented the lowest genetic diversity, the highest levels of inbreeding and the smallest effective population size. ADMIXTURE analysis also demonstrated that ducks were clearly divided into these three populations at K = 3. Selection signals in the commercial population were associated with growth and muscle development pathways, such as the mTOR signaling pathway and ErbB signaling pathway, and two key genomic regions (Chr1: 70.25 to 74.00 Mb and Chr2: 97.10 to 99.76 Mb) containing important genes, such as LRP6, BORCS5, and EDN1, were identified. In contrast, selection signals in the local population were associated with immune-related pathways involving NCAM2 and MPHOSPH6. Furthermore, PTGS2 and PLA2G4A genes were positively selected in egg breeds, whereas KCNK16, KCNK5, and KCNK17 genes were in dual-purpose breeds. CONCLUSION Because of artificial selection, wild, local and commercial populations presented obvious genetic differences. The selection signal analysis revealed that LRP6, BORCS5, and EDN1 are important for growth and muscle development; NCAM2 and MPHOSPH6 are for immune traits; and PTGS2 and PLA2G4A are for egg-related traits.
Collapse
Affiliation(s)
- Zhirong Huang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| | - Liyun Zhang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| | - Maojun Luo
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| | - Xumeng Zhang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| | - Yunmao Huang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| | - Yunbo Tian
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| | - Zhongping Wu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| | - Xiujin Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou,
China
| |
Collapse
|
2
|
Chen G, Liu J, Guo Y, Sun P. Mechanisms for Regulatory Effects of Exercise on Metabolic Diseases from the Lactate-Lactylation Perspective. Int J Mol Sci 2025; 26:3469. [PMID: 40331975 PMCID: PMC12027343 DOI: 10.3390/ijms26083469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic diseases, including cardiovascular diseases, type 2 diabetes mellitus (T2DM), osteoporosis, and non-alcoholic fatty liver disease (NAFLD), constitute a major global health burden associated with chronic morbidity and mortality. Lactate, once considered as a metabolic byproduct, has emerged as a key regulator of cellular reprogramming through lactylation, a novel post-translational modification (PTM) that dynamically couples metabolic flux to chromatin remodeling. Lactylation exerts dual regulatory roles as a signaling molecule via GPR81/GPR4-mediated pathways and as a substrate for the covalent modification of histones and metabolic enzymes. Pathologically, chronic hyperlactatemia suppresses mitochondrial biogenesis, driving metabolic cardiomyopathy through the epigenetic silencing of oxidative metabolism genes. Conversely, exercise-induced lactate surges transiently enhance insulin sensitivity via AMPK/PGC-1α/GLUT4 signaling, resolve inflammation through GPR81-mediated M2 macrophage polarization, and restore mitochondrial function via lactylation-dependent pathways. This review delineates lactylation as a spatiotemporal rheostat: chronic dysregulation perpetuates metabolic disorders, whereas acute exercise-mediated lactylation remodels transcriptional networks to restore metabolic homeostasis. Future research should integrate multiomics to clarify lactylation's spatiotemporal dynamics, tissue-specific thresholds, metabolism-immunity interactions, and metabolic-epigenetic crosstalk for the precision management of metabolic diseases.
Collapse
Affiliation(s)
- Guannan Chen
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
| | - Jinchao Liu
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
| | - Yilan Guo
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; (G.C.); (J.L.); (Y.G.)
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai 200241, China
| |
Collapse
|
3
|
He Y, Liu T, Peng X, Yao C, Zhou D, Song C, Wei Z, Chen J, Liu Z, Jiang F. Molecular mechanism of mitochondrial autophagy mediating impaired energy metabolism leading to osteoporosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167685. [PMID: 39842521 DOI: 10.1016/j.bbadis.2025.167685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Osteoporosis (OP) is a bone metabolic disease caused by decreased bone mass leading to destruction of bone microstructure. Treatment of OP is characterized by a lifelong nature, causing extreme financial and psychological burdens to patients. Hormonal abnormalities, cellular autophagy, Ferroptosis, and oxidative stress are all part of the intricate and varied pathophysiology of OP. Recent research has revealed that mitochondrial dysfunction is a significant factor in the onset and progression of OP. By regulating bone marrow mesenchymal stem cell differentiation through various signaling pathways and cytokines, abnormal mitochondrial energy metabolism brought on by oxidative stress processes impacts osteoblast and osteoclast proliferation and differentiation, causing an imbalance in bone metabolism that ultimately results in OP. Therefore, one possible method to prevent and manage OP may be to use mitochondria as a carrier to trigger osteogenic differentiation of bone marrow mesenchymal stem cells from mitochondrial energy consumption, oxidative stress, autophagy, and osteoclast death. In order to offer some theoretical references and therapeutic approaches for the clinical prevention and treatment of OP, we will examine the pathophysiology of OP from mitochondrial dysfunction in this work.
Collapse
Affiliation(s)
- Yuheng He
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Tao Liu
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xin Peng
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chaorui Yao
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; College of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Daqian Zhou
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Chao Song
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Department of Orthopedics, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Zhangchao Wei
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jinwen Chen
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Zongchao Liu
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Luzhou Longmatan District People's Hospital, Luzhou 646000, Sichuan Province, China.
| | - Feng Jiang
- Department of Orthopedics and Traumatology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
4
|
Lin B, Liu H, Liu H, Su L, Sun K, Feng H, Liu Y, Yu M, Han D. A novel WNT10A variant impairs the homeostasis of alveolar bone mesenchymal stem cells. Oral Dis 2025; 31:168-180. [PMID: 38852166 DOI: 10.1111/odi.15032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
OBJECTIVES To explore the influence of a novel WNT10A variant on bone mineral density, proliferation, and osteogenic differentiation capacities of alveolar bone mesenchymal stem cells in humans. SUBJECTS AND METHODS Whole-exome sequencing and Sanger sequencing were utilized to detect gene variants in a family with non-syndromic tooth agenesis (NSTA). The panoramic mandibular index was calculated on the proband with WNT10A variant and normal controls to evaluate bone mineral density. Alveolar bone mesenchymal stem cells from the proband with a novel WNT10A variant and normal controls were isolated and cultured, then proliferation and osteogenic differentiation capacities were evaluated and compared. RESULTS We identified a novel WNT10A pathogenic missense variant (c.353A > G/p. Tyr118Cys) in a family with NSTA. The panoramic mandibular index of the proband implied a reduction in bone mineral density. Moreover, the proliferation and osteogenic differentiation capacities of alveolar bone mesenchymal stem cells from the proband with WNT10A Tyr118Cys variant were significantly decreased. CONCLUSIONS Our findings broaden the spectrum of WNT10A variants in patients with non-syndromic oligodontia, suggest an association between WNT10A and the proliferation and osteogenic differentiation of alveolar bone mesenchymal stem cells, and demonstrate that WNT10A is involved in maintaining jaw bone homeostasis.
Collapse
Affiliation(s)
- Bichen Lin
- Frist Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Haochen Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Hangbo Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Lanxin Su
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Kai Sun
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Hailan Feng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yang Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Miao Yu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Dong Han
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
5
|
Song F, Marmo T, Song C, Liao X, Long F. Wnt7b overexpression in osteoblasts stimulates bone formation and reduces obesity in mice on a high-fat diet. JBMR Plus 2024; 8:ziae122. [PMID: 39434845 PMCID: PMC11491285 DOI: 10.1093/jbmrpl/ziae122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 10/23/2024] Open
Abstract
Previous studies have shown that Wnt7b potently stimulates bone formation by promoting osteoblast differentiation and activity. As high-fat feeding leads to obesity and systemic metabolic dysregulation, here we investigate the potential benefit of Wnt7b overexpression in osteoblasts on both bone and whole-body metabolism in mice fed with a high-fat diet (HFD). Wnt7b overexpression elicited massive overgrowth of trabecular and cortical bone but seemed to ameliorate body fat accumulation in mice with prolonged HFD feeding. In addition, Wnt7b overexpression modestly improved glucose tolerance in male mice on HFD. Collectively, the results indicate that targeted overexpression of Wnt7b in osteoblasts not only stimulates bone formation but also improves certain aspects of global metabolism in overnourished mice.
Collapse
Affiliation(s)
- Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Tyler Marmo
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Chao Song
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Xueyang Liao
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, United States
| |
Collapse
|
6
|
Bertels JC, He G, Long F. Metabolic reprogramming in skeletal cell differentiation. Bone Res 2024; 12:57. [PMID: 39394187 PMCID: PMC11470040 DOI: 10.1038/s41413-024-00374-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/13/2024] Open
Abstract
The human skeleton is a multifunctional organ made up of multiple cell types working in concert to maintain bone and mineral homeostasis and to perform critical mechanical and endocrine functions. From the beginning steps of chondrogenesis that prefigures most of the skeleton, to the rapid bone accrual during skeletal growth, followed by bone remodeling of the mature skeleton, cell differentiation is integral to skeletal health. While growth factors and nuclear proteins that influence skeletal cell differentiation have been extensively studied, the role of cellular metabolism is just beginning to be uncovered. Besides energy production, metabolic pathways have been shown to exert epigenetic regulation via key metabolites to influence cell fate in both cancerous and normal tissues. In this review, we will assess the role of growth factors and transcription factors in reprogramming cellular metabolism to meet the energetic and biosynthetic needs of chondrocytes, osteoblasts, or osteoclasts. We will also summarize the emerging evidence linking metabolic changes to epigenetic modifications during skeletal cell differentiation.
Collapse
Affiliation(s)
- Joshua C Bertels
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Guangxu He
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Orthopedics, The Second Xiangya Hospital, Changsha, Hunan, China
| | - Fanxin Long
- Department of Surgery, Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
7
|
You C, Shen F, Yang P, Cui J, Ren Q, Liu M, Hu Y, Li B, Ye L, Shi Y. O-GlcNAcylation mediates Wnt-stimulated bone formation by rewiring aerobic glycolysis. EMBO Rep 2024; 25:4465-4487. [PMID: 39256595 PMCID: PMC11467389 DOI: 10.1038/s44319-024-00237-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Wnt signaling is an important target for anabolic therapies in osteoporosis. A sclerostin-neutralizing antibody (Scl-Ab), that blocks the Wnt signaling inhibitor (sclerostin), has been shown to promote bone mass in animal models and clinical studies. However, the cellular mechanisms by which Wnt signaling promotes osteogenesis remain to be further investigated. O-GlcNAcylation, a dynamic post-translational modification of proteins, controls multiple critical biological processes including transcription, translation, and cell fate determination. Here, we report that Wnt3a either induces O-GlcNAcylation rapidly via the Ca2+-PKA-Gfat1 axis, or increases it in a Wnt-β-catenin-dependent manner following prolonged stimulation. Importantly, we find O-GlcNAcylation indispensable for osteoblastogenesis both in vivo and in vitro. Genetic ablation of O-GlcNAcylation in the osteoblast-lineage diminishes bone formation and delays bone fracture healing in response to Wnt stimulation in vivo. Mechanistically, Wnt3a induces O-GlcNAcylation at Serine 174 of PDK1 to stabilize the protein, resulting in increased glycolysis and osteogenesis. These findings highlight O-GlcNAcylation as an important mechanism regulating Wnt-induced glucose metabolism and bone anabolism.
Collapse
Affiliation(s)
- Chengjia You
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fangyuan Shen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Puying Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingyao Cui
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiaoyue Ren
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Moyu Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujie Hu
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Boer Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Yu Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
9
|
Stegen S, Carmeliet G. Metabolic regulation of skeletal cell fate and function. Nat Rev Endocrinol 2024; 20:399-413. [PMID: 38499689 DOI: 10.1038/s41574-024-00969-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2024] [Indexed: 03/20/2024]
Abstract
Bone development and bone remodelling during adult life are highly anabolic processes requiring an adequate supply of oxygen and nutrients. Bone-forming osteoblasts and bone-resorbing osteoclasts interact closely to preserve bone mass and architecture and are often located close to blood vessels. Chondrocytes within the developing growth plate ensure that bone lengthening occurs before puberty, but these cells function in an avascular environment. With ageing, numerous bone marrow adipocytes appear, often with negative effects on bone properties. Many studies have now indicated that skeletal cells have specific metabolic profiles that correspond to the nutritional microenvironment and their stage-specific functions. These metabolic networks provide not only skeletal cells with sufficient energy, but also biosynthetic intermediates that are necessary for proliferation and extracellular matrix synthesis. Moreover, these metabolic pathways control redox homeostasis to avoid oxidative stress and safeguard cell survival. Finally, several intracellular metabolites regulate the activity of epigenetic enzymes and thus control the fate and function of skeletal cells. The metabolic profile of skeletal cells therefore not only reflects their cellular state, but can also drive cellular activity. Insight into skeletal cell metabolism will thus not only advance our understanding of skeletal development and homeostasis, but also of skeletal disorders, such as osteoarthritis, diabetic bone disease and bone malignancies.
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium.
| |
Collapse
|
10
|
Kaur S, Kumari P, Singh G, Joshi N, Kaur T, Dhiman V, Singh G, Sachdeva N, Kumar D, Barnwal RP, Bhadada SK. Unveiling novel metabolic alterations in postmenopausal osteoporosis and type 2 diabetes mellitus through NMR-based metabolomics: A pioneering approach for identifying early diagnostic markers. J Proteomics 2024; 302:105200. [PMID: 38772440 DOI: 10.1016/j.jprot.2024.105200] [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: 04/26/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND AND AIMS Postmenopausal osteoporosis (PMO) and type 2 diabetes mellitus (T2DM) frequently coexist in postmenopausal women. The study aimed to explore metabolic variations linked to these circumstances and their simultaneous presence through proton nuclear magnetic resonance metabolomics (1H NMR). MATERIALS AND METHODS Serum samples from 80 postmenopausal women, including 20 PMO individuals, 20 T2DM, 20 T2DM + PMO, and 20 healthy postmenopausal women, were analyzed using 1H NMR spectroscopy. RESULTS Our study revealed significant metabolic profile differences among the four groups. Notably, the T2DM + PMO group showed elevated levels of alanine, pyruvate, glutamate, lactate, and aspartate, indicating their involvement in lipid metabolism, energy, and amino acids. Importantly, our multivariate statistical analysis identified a metabolite set that accurately distinguished the groups, suggesting its potential as an early diagnostic marker. CONCLUSION The 1H NMR metabolomics approach uncovered metabolic biomarkers intricately linked to postmenopausal osteoporosis (PMO), type 2 diabetes mellitus (T2DM), and their concurrent presence. Among these biomarkers, alanine emerged as a pivotal player, showing its significant role in the metabolic landscape associated with PMO and T2DM. These findings shed light on the pathophysiological mechanisms underlying these conditions and underscore alanine's potential as a diagnostic biomarker.
Collapse
Affiliation(s)
- Simran Kaur
- Department of Biophysics, Panjab University, Chandigarh, India; Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Poonam Kumari
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Gurvinder Singh
- Centre of Biomedical Research, SGPGIMS campus, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Nainesh Joshi
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Takdeer Kaur
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Vandana Dhiman
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Naresh Sachdeva
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Dinesh Kumar
- Centre of Biomedical Research, SGPGIMS campus, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | | | - Sanjay Kumar Bhadada
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India.
| |
Collapse
|
11
|
Zhou F, Wang Z, Zhang G, Wu Y, Xiong Y. Immunosenescence and inflammaging: Conspiracies against alveolar bone turnover. Oral Dis 2024; 30:1806-1817. [PMID: 37288702 DOI: 10.1111/odi.14642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/11/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE Inflammaging and immunosenescence are characteristics of senescent immune system alterations. This review provides insights into inflammaging and immunosenescence in periodontitis and focuses on the innerlink of inflammaging and immunosenescence in alveolar bone turnover from a perspective of cell-cell interaction. METHODS This review is conducted by a narrative approach to discuss the effect of inflammaging and immunosenescence in aging-related alveolar bone loss. A comprehensive literature research in PubMed and Google was applied to identify reports in English. RESULTS Inflammaging is concerned with abnormal M1 polarization and increasing circulating inflammatory cytokines, while immunosenescence involves reduced infection and vaccine responses, depressed antimicrobial function, and infiltration of aged B cells and memory T cells. TLR-mediated inflammaging and altered adaptive immunity significantly affect alveolar bone turnover and aggravate aging-related alveolar bone loss. Besides, energy consumption also plays a vital role in aged immune and skeletal system of periodontitis. CONCLUSIONS Senescent immune system exerts a significant function in aging-related alveolar bone loss. Inflammaging and immunosenescence interact functionally and mechanistically, which affects alveolar bone turnover. Therefore, further clinical treatment strategies targeting alveolar bone loss could be based on the specific molecular mechanism connecting inflammaging, immunosenescence, and alveolar bone turnover.
Collapse
Affiliation(s)
- Feng Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhanqi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guorui Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yingying Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Huang X, Meng H, Shou Z, Zhou H, Chen L, Yu J, Hu K, Bai Z, Chen C. Machine learning-mediated identification of ferroptosis-related genes in osteonecrosis of the femoral head. FEBS Open Bio 2024; 14:455-465. [PMID: 38212150 PMCID: PMC10909969 DOI: 10.1002/2211-5463.13764] [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: 07/14/2023] [Revised: 10/18/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a condition caused by a disruption or damage to the femoral head's blood supply, which causes the death of bone cells and bone marrow components and prevents future regeneration. Ferroptosis, a type of controlled cell death, is caused by iron-dependent lipid peroxidation. Here, we identified ferroptosis-related genes and infiltrating immune cells involved in ONFH and predicted the underlying molecular mechanisms. The GSE123568 dataset was subjected to differential expression analysis to identify genes related to ferroptosis. Subsequently, GO and KEGG pathway enrichment analyses, as well as protein-protein interaction (PPI) network analysis, were conducted. Hub genes involved in ferroptosis were identified using machine learning and other techniques. Additionally, immune infiltration analysis and lncRNA-miRNA-mRNA network prediction analysis were performed. Finally, we determined whether ferroptosis occurred by measuring iron content. The hub genes were validated by ROC curve analysis and qRT-PCR. Four ferroptosis-related hub genes (MAPK3, PTGS2, STK11, and SLC2A1) were identified. Additionally, immune infiltration analysis revealed a strong correlation among ONFH, hub genes, and various immune cells. Finally, we predicted the network relationship between differentially expressed lncRNAs and hub genes in the lncRNA-miRNA-mRNA network. MAPK3, PTGS2, STK11, and SLC2A1 have been identified as potential ferroptosis-related biomarkers and drug targets for the diagnosis and prognosis of ONFH, while some immune cells, as well as the interaction between lncRNA, miRNA, and mRNA, have also been identified as potential pathogenesis markers and therapeutic targets.
Collapse
Affiliation(s)
- Xiaojing Huang
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
| | - Hongming Meng
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Wenzhou Medical UniversityChina
| | - Zeyu Shou
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Wenzhou Medical UniversityChina
| | - Han Zhou
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Wenzhou Medical UniversityChina
| | - Liangyan Chen
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Wenzhou Medical UniversityChina
| | - Jiahuan Yu
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Wenzhou Medical UniversityChina
| | - Kai Hu
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Wenzhou Medical UniversityChina
| | - Zhibiao Bai
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
| | - Chun Chen
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical UniversityChina
- Wenzhou Medical UniversityChina
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang ProvinceWenzhouChina
- Zhejiang Engineering Research Center for Hospital Emergency and Process DigitizationWenzhouChina
| |
Collapse
|
13
|
Lu H, Fan Y, Yan Q, Chen Z, Wei Z, Liu Y, Zhang J, Huang Z, Fang H, Zhou C, Chen Z. Identification and validation of ferroptosis-related biomarkers in steroid-induced osteonecrosis of the femoral head. Int Immunopharmacol 2023; 124:110906. [PMID: 37690237 DOI: 10.1016/j.intimp.2023.110906] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
OBJECTIVES Treatment of steroid-induced osteonecrosis of the femoral head (SIONFH) is challenging. Due to the limited understanding of its molecular mechanisms, investigating the potential mechanisms of ferroptosis will shed light on SIONFH and provide directions for treating this disease. METHODS The GSE123568 dataset was utilized to apply various bioinformatics methodologies to identify ferroptosis-related hub genes (FRHGs). Subsequently, the importance of these genes and the reliability of the results were confirmed using protein data-independent acquisition (DIA) and cell experiments. Finally, we assessed the correlation between FRHG expression and immune cell infiltration. RESULTS Thirty-one hub genes were identified and validated by constructing a protein-protein interaction network and subsequent screening using experimentally determined interactions. These 31 hub genes were enriched in immunity, the AMPK signaling pathway, and the Toll-like receptor signaling pathway. Next, we identified a diagnostic marker comprising two ferroptosis-related genes, NCF2 and SLC2A1. The differential expression of these two genes in healthy and necrotic regions was confirmed by protein DIA analysis. Cell experiments verified the link between FRHGs and ferroptosis and preliminarily explored the potential mechanism of the antioxidant vitexin in promoting osteogenic differentiation in cells. The diagnostic efficiency of these two markers was confirmed by receiver operating characteristic curve (ROC) curves, yielding an area under the curve of 1.0. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) indicated enrichment of FRHGs in the superoxide anion and HIF-1 signaling pathways. A significant correlation was observed between FRHGs and various immune cell populations. CONCLUSION NCF2 and SLC2A1 are promising ferroptosis-related diagnostic biomarkers of SIONFH. Concurrently, we embarked on a preliminary investigation to elucidate the potential mechanism underlying the promotion of osteogenic differentiation by the antioxidant vitexin. Moreover, these biomarkers are associated with distinct immune cell populations.
Collapse
Affiliation(s)
- Hongduo Lu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Yinuo Fan
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China; The Third Clinical Medical College, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Qian Yan
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Zhiwen Chen
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Zhiming Wei
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Yuhao Liu
- The Department of Orthopedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Jiahao Zhang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Zeqing Huang
- The Department of Orthopedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Hanjun Fang
- The Department of Orthopedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Chi Zhou
- The Department of Orthopedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| | - Zhenqiu Chen
- The Department of Orthopedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun District, Guangzhou 510405, Guangdong Province, China.
| |
Collapse
|
14
|
Sabini E, Arboit L, Khan MP, Lanzolla G, Schipani E. Oxidative phosphorylation in bone cells. Bone Rep 2023; 18:101688. [PMID: 37275785 PMCID: PMC10238578 DOI: 10.1016/j.bonr.2023.101688] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
The role of energy metabolism in bone cells is an active field of investigation. Bone cells are metabolically very active and require high levels of energy in the form of adenosine triphosphate (ATP) to support their function. ATP is generated in the cytosol via glycolysis coupled with lactic acid fermentation and in the mitochondria via oxidative phosphorylation (OXPHOS). OXPHOS is the final convergent metabolic pathway for all oxidative steps of dietary nutrients catabolism. The formation of ATP is driven by an electrochemical gradient that forms across the mitochondrial inner membrane through to the activity of the electron transport chain (ETC) complexes and requires the presence of oxygen as the final electron acceptor. The current literature supports a model in which glycolysis is the main source of energy in undifferentiated mesenchymal progenitors and terminally differentiated osteoblasts, whereas OXPHOS appears relevant in an intermediate stage of differentiation of those cells. Conversely, osteoclasts progressively increase OXPHOS during differentiation until they become multinucleated and mitochondrial-rich terminal differentiated cells. Despite the abundance of mitochondria, mature osteoclasts are considered ATP-depleted, and the availability of ATP is a critical factor that regulates the low survival capacity of these cells, which rapidly undergo death by apoptosis. In addition to ATP, bioenergetic metabolism generates reactive oxygen species (ROS) and intermediate metabolites that regulate a variety of cellular functions, including epigenetics changes of genomic DNA and histones. This review will briefly discuss the role of OXPHOS and the cross-talks OXPHOS-glycolysis in the differentiation process of bone cells.
Collapse
Affiliation(s)
- Elena Sabini
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Lorenzo Arboit
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Giulia Lanzolla
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia 19104, PA, USA
| |
Collapse
|
15
|
Song F, Lee WD, Marmo T, Ji X, Song C, Liao X, Seeley R, Yao L, Liu H, Long F. Osteoblast-intrinsic defect in glucose metabolism impairs bone formation in type II diabetic male mice. eLife 2023; 12:e85714. [PMID: 37144869 PMCID: PMC10198725 DOI: 10.7554/elife.85714] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/04/2023] [Indexed: 05/06/2023] Open
Abstract
Skeletal fragility is associated with type 2 diabetes mellitus (T2D), but the underlying mechanism is not well understood. Here, in a mouse model for youth-onset T2D, we show that both trabecular and cortical bone mass is reduced due to diminished osteoblast activity. Stable isotope tracing in vivo with 13C-glucose demonstrates that both glycolysis and glucose fueling of the TCA cycle are impaired in diabetic bones. Similarly, Seahorse assays show suppression of both glycolysis and oxidative phosphorylation by diabetes in bone marrow mesenchymal cells as a whole, whereas single-cell RNA sequencing reveals distinct modes of metabolic dysregulation among the subpopulations. Metformin not only promotes glycolysis and osteoblast differentiation in vitro, but also improves bone mass in diabetic mice. Finally, osteoblast-specific overexpression of either Hif1a, a general inducer of glycolysis, or Pfkfb3 which stimulates a specific step in glycolysis, averts bone loss in T2D mice. The study identifies osteoblast-intrinsic defects in glucose metabolism as an underlying cause of diabetic osteopenia, which may be targeted therapeutically.
Collapse
Affiliation(s)
- Fangfang Song
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan UniversityWuhanChina
| | - Won Dong Lee
- Lewis Sigler Institute for Integrative Genomics, Princeton UniversityPrincetonUnited States
| | - Tyler Marmo
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Xing Ji
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Chao Song
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Xueyang Liao
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Rebecca Seeley
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Lutian Yao
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Haoran Liu
- Department of Computer Science, New Jersey Institute of TechnologyNewarkUnited States
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Deaprtment of Orthopedic Surgery, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
16
|
Zhu Z, Gao S, Zhu H, Chen Y, Wu D, Chen Z, Huang Y, Wu X, Hu N, Chen D, Huang W, Chen H. Metformin improves fibroblast metabolism and ameliorates arthrofibrosis in rats. J Orthop Translat 2023; 40:92-103. [PMID: 37457314 PMCID: PMC10338908 DOI: 10.1016/j.jot.2023.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/03/2023] [Accepted: 05/30/2023] [Indexed: 07/18/2023] Open
Abstract
Background Emerging studies have suggested an essential role of fibroblast metabolic reprogramming in the pathogenesis of arthrofibrosis. The metabolic modulator metformin appears to be a therapeutic candidate for fibrotic disorders. However, whether metformin could alleviate arthrofibrosis has not been defined. In this study we have determined if treatment with metformin has beneficial effect on arthrofibrosis and its underlying mechanism. Methods Articular capsule samples were collected from patients with/without arthrofibrosis to perform gene and protein expression analysis. Arthrofibrosis animal model was established to examine the anti-fibrotic effect of metformin. Cell culture experiments were conducted to determine the mechanism by which metformin inhibits fibroblast activation. Results We found that glycolysis was upregulated in human fibrotic articular capsules. In an arthrofibrosis animal model, intra-articular injection of metformin mitigated inflammatory reactions, downregulated expression of both fibrotic and glycolytic markers, improved range of motion (ROM) of the joint, and reduced capsular fibrosis and thickening. At the cellular level, metformin inhibited the activation of fibroblasts and mitigated the abundant influx of glucose into activated fibroblasts. Interestingly, metformin prompted a metabolic shift from oxidative phosphorylation to aerobic glycolysis in activated fibroblasts, resulting in the anti-fibrotic effect of metformin. Conclusion Metformin decreased glycolysis, causing a metabolic shift toward aerobic glycolysis in activated fibroblasts and has beneficial effect on the treatment of arthrofibrosis.The translational potential of this article: The findings of this study demonstrated the therapeutic effect of metformin on arthrofibrosis and defined novel targets for the treatment of articular fibrotic disorders.
Collapse
Affiliation(s)
- Zhenglin Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Shengqiang Gao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Hui Zhu
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Chen
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dandong Wu
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyu Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Xiangdong Wu
- Department of Orthopaedic Surgery, Peking University Fourth School of Clinical Medicine/Beijing Jishuitan Hospital, Beijing, China
| | - Ning Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Di Chen
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institue of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Bioinformatics-Based Analysis of Key Genes in Steroid-Induced Osteonecrosis of the Femoral Head That Are Associated with Copper Metabolism. Biomedicines 2023; 11:biomedicines11030873. [PMID: 36979852 PMCID: PMC10045807 DOI: 10.3390/biomedicines11030873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a common disabling disease. Copper has positive effects on cells that regulate bone metabolism. However, the relationship between copper metabolism (CM) and steroid-induced ONFH (SONFH) remains unclear. The GSE123568 dataset was downloaded from the Gene Expression Omnibus. The differentially expressed CM-related SONFH genes (DE-CMR-SONFHGs) were identified via differential analysis and weighted gene coexpression network analysis (WGCNA). Receiver operating characteristic (ROC) analysis was performed for the predictive accuracy of key genes. Targeting drugs and the copper death-related genes (CDRGs) relevant to key genes were investigated. The bioinformatics results were confirmed via quantitative real-time polymerase chain reaction (qRT–PCR) and Western blot (WB) analysis. Two out of 106 DE-CMR-SONFHGs were identified as key genes (PNP and SLC2A1), which had diagnostic value in distinguishing SONFH from control samples and were related to various immune cell infiltrations. Eleven PMP-targeting drugs and five SLC2A1-targeting drugs were identified. The qRT–PCR, as well as WB, results confirmed the downregulation PNP and SLC2A1 and high expression of the CDRGs DLD, PDHB, and MTF1, which are closely related to these two key genes. In conclusion, PNP and SLC2A1 were identified as key genes related to SONFH and may provide insights for SONFH treatment.
Collapse
|
18
|
Zhu Z, Chen Y, Zou J, Gao S, Wu D, Li X, Hu N, Zhao J, Huang W, Chen H. Lactate Mediates the Bone Anabolic Effect of High-Intensity Interval Training by Inducing Osteoblast Differentiation. J Bone Joint Surg Am 2023; 105:369-379. [PMID: 36728458 DOI: 10.2106/jbjs.22.01028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND High-intensity interval training (HIIT) reportedly improves bone metabolism and increases bone mineral density (BMD). The purpose of the present study was to investigate whether lactate mediates the beneficial effects of exercise on BMD, bone microarchitecture, and biomechanical properties in an established osteoporotic animal model. In addition, we hypothesized that lactate-induced bone augmentation is achieved through enhanced osteoblast differentiation and mineralization. METHODS A total of 50 female C57BL/6 mice were randomly allocated into 5 groups: the nonovariectomized group, the ovariectomized group (OVX), the HIIT group (OVX + HIIT), the HIIT with lactate transporter inhibition group (OVX + HIIT + INH), and the lactate subcutaneous injection group (OVX + LAC). After 7 weeks of intervention, bone mass, bone strength, and bone formation/resorption processes were evaluated via microcomputed tomography (micro-CT), biomechanical testing, histological analysis, and serum biochemical assays; in vitro studies were performed to explore the bone anabolic effect of lactate at the cellular level. RESULTS Micro-CT revealed significantly increased BMD in both the OVX + HIIT group (mean difference, 41.03 mg hydroxyapatite [HA]/cm 3 [95% CI, 2.51 to 79.54 mg HA/cm 3 ]; p = 0.029) and the OVX + LAC group (mean difference, 40.40 mg HA/cm 3 [95% CI, 4.08 to 76.71 mg HA/cm 3 ]; p = 0.031) compared with the OVX group. Biomechanical testing demonstrated significantly improved mechanical properties in those 2 groups. However, the beneficial effects of exercise on bone microstructure and biomechanics were largely abolished by blocking the lactate transporter. Notably, histological and biochemical results indicated that increased bone formation was responsible for the bone augmentation effects of HIIT and lactate. Cell culture studies showed a marked increase in the expression of osteoblastic markers with lactate treatment, which could be eliminated by blocking the lactate transporter. CONCLUSIONS Lactate may have mediated the bone anabolic effect of HIIT in osteoporotic mice, which may have resulted from enhanced osteoblast differentiation and mineralization. CLINICAL RELEVANCE Lactate may mediate the bone anabolic effect of HIIT and serve as a potential inexpensive therapeutic strategy for bone augmentation.
Collapse
Affiliation(s)
- Zhenglin Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yi Chen
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jing Zou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Shengqiang Gao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dandong Wu
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xuelun Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ning Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Orthopedic Laboratory of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
19
|
Song F, Lee WD, Marmo T, Ji X, Song C, Liao X, Seeley R, Yao L, Liu H, Long F. Osteoblast-intrinsic defect in glucose metabolism impairs bone formation in type II diabetic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524248. [PMID: 36711657 PMCID: PMC9882117 DOI: 10.1101/2023.01.16.524248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Skeletal fragility is associated with type 2 diabetes mellitus (T2D), but the underlying mechanism is not well understood. Here, in a mouse model for youth-onset T2D, we show that both trabecular and cortical bone mass are reduced due to diminished osteoblast activity. Stable isotope tracing in vivo with 13 C-glucose demonstrates that both glycolysis and glucose fueling of the TCA cycle are impaired in diabetic bones. Similarly, Seahorse assays show suppression of both glycolysis and oxidative phosphorylation by diabetes in bone marrow mesenchymal cells as a whole, whereas single-cell RNA sequencing reveals distinct modes of metabolic dysregulation among the subpopulations. Metformin not only promotes glycolysis and osteoblast differentiation in vitro, but also improves bone mass in diabetic mice. Finally, targeted overexpression of Hif1a or Pfkfb3 in osteoblasts of T2D mice averts bone loss. The study identifies osteoblast-intrinsic defects in glucose metabolism as an underlying cause of diabetic osteopenia, which may be targeted therapeutically.
Collapse
Affiliation(s)
- Fangfang Song
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Won Dong Lee
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Tyler Marmo
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
| | - Xing Ji
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
| | - Chao Song
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
| | - Xueyang Liao
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
| | - Rebbeca Seeley
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
| | - Lutian Yao
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
| | - Haoran Liu
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia
- Deaprtment of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
20
|
Li Z, Yue M, Liu X, Liu Y, Lv L, Zhang P, Zhou Y. The PCK2-glycolysis axis assists three-dimensional-stiffness maintaining stem cell osteogenesis. Bioact Mater 2022; 18:492-506. [PMID: 35415308 PMCID: PMC8971594 DOI: 10.1016/j.bioactmat.2022.03.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 01/02/2023] Open
Abstract
Understanding mechanisms underlying the heterogeneity of multipotent stem cells offers invaluable insights into biogenesis and tissue development. Extracellular matrix (ECM) stiffness has been acknowledged as a crucial factor regulating stem cell fate. However, how cells sense stiffness cues and adapt their metabolism activity is still unknown. Here we report the novel role of mitochondrial phosphoenolpyruvate carboxykinase (PCK2) in enhancing osteogenesis in 3D ECM via glycolysis. We experimentally mimicked the physical characteristics of 3D trabeculae network of normal and osteoporotic bone with different microstructure and stiffness, observing that PCK2 promotes osteogenesis in 3D ECM with tunable stiffness in vitro and in vivo. Mechanistically, PCK2 enhances the rate-limiting metabolic enzyme pallet isoform phosphofructokinase (PFKP) in 3D ECM, and further activates AKT/extracellular signal-regulated kinase 1/2 (ERK1/2) cascades, which directly regulates osteogenic differentiation of MSCs. Collectively, our findings implicate an intricate crosstalk between cell mechanics and metabolism, and provide new perspectives for strategies of osteoporosis. As the key rate-limiting enzyme of gluconeogenesis, PCK2 manipulates osteogenesis in stiff and soft ECM in vitro and in vivo. PCK2 regulates osteogenic capacity of BMMSCs in 3D ECM with different stiffness, via modulating glycolysis and regulating PFKP-AKT/ERK signaling pathways.
Collapse
Affiliation(s)
- Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Muxin Yue
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Corresponding author. Vice Professor of Department of Prosthodontics, School and Hospital of Stomatology of Peking University, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Corresponding author. President of School and Hospital of Stomatology of Peking University, Professor of Department of Prosthodontics, Vice-Director for National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Vice-Director for the National Clinical Research Center of Oral Diseases (PKU), 22 Zhongguancun South Avenue, Haidian District, Beijing, 10081, PR China.
| |
Collapse
|
21
|
Abstract
The mammalian skeleton is integral to whole body physiology with a multitude of functions beyond mechanical support and locomotion, including support of hematopoiesis, mineral homeostasis and potentially other endocrine roles. Formation of the skeleton begins in the embryo and mostly from a cartilage template that is ultimately replaced by bone through endochondrial ossification. Skeletal development and maturation continue after birth in most species and last into the second decade of postnatal life in humans. In the mature skeleton, articular cartilage lining the synovial joint surfaces is vital for bodily movement and damages to the cartilage are a hallmark of osteoarthritis. The mature bone tissue undergoes continuous remodeling initiated with bone resorption by osteoclasts and completed with bone formation from osteoblasts. In a healthy state, the exquisite balance between bone resorption and formation is responsible for maintaining a stable bone mass and structural integrity, while meeting the physiological needs for minerals via controlled release from bone. Disruption of the balance in favor of bone resorption is the root cause for osteoporosis. Whereas osteoclasts pump molar quantities of hydrochloric acid to dissolve the bone minerals in a process requiring ATP hydrolysis, osteoblasts build bone mass by synthesizing and secreting copious amounts of bone matrix proteins. Thus, both osteoclasts and osteoblasts engage in energy-intensive activities to fulfill their physiological functions, but the bioenergetics of those and other skeletal cell types are not well understood. Nonetheless, the past ten years have witnessed a resurgence of interest in studies of skeletal cell metabolism, resulting in an unprecedented understanding of energy substrate utilization and its role in cell fate and activity regulation. The present review attempts to synthesize the current findings of glucose metabolism in chondrocytes, osteoblasts and osteoclasts. Advances with the other relevant cell types including skeletal stem cells and marrow adipocytes will not be discussed here as they have been extensively reviewed recently by others (van Gastel and Carmeliet, 2021). Elucidation of the bioenergetic mechanisms in the skeletal cells is likely to open new avenues for developing additional safe and effective bone therapies.
Collapse
Affiliation(s)
- Fanxin Long
- Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, Department of Orthopedic Surgery, University of Pennsylvania, United States of America
| |
Collapse
|
22
|
Weber DR, Long F, Zemel BS, Kindler JM. Glycemic Control and Bone in Diabetes. Curr Osteoporos Rep 2022; 20:379-388. [PMID: 36214991 PMCID: PMC9549036 DOI: 10.1007/s11914-022-00747-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2022] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW This review summarizes recent developments on the effects of glycemic control and diabetes on bone health. We discuss the foundational cellular mechanisms through which diabetes and impaired glucose control impact bone biology, and how these processes contribute to bone fragility in diabetes. RECENT FINDINGS Glucose is important for osteoblast differentiation and energy consumption of mature osteoblasts. The role of insulin is less clear, but insulin receptor deletion in mouse osteoblasts reduces bone formation. Epidemiologically, type 1 (T1D) and type 2 diabetes (T2D) associate with increased fracture risk, which is greater among people with T1D. Accumulation of cortical bone micro-pores, micro-vascular complications, and AGEs likely contribute to diabetes-related bone fragility. The effects of youth-onset T2D on peak bone mass attainment and subsequent skeletal fragility are of particular concern. Further research is needed to understand the effects of hyperglycemia on skeletal health through the lifecycle, including the related factors of inflammation and microvascular damage.
Collapse
Affiliation(s)
- David R Weber
- Division of Endocrinology and Diabetes, Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia,, PA, USA
| | - Fanxin Long
- Department of Orthopedic Surgery, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Babette S Zemel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Division of GI, Hepatology & Nutrition, Roberts Center for Pediatric Research, 2716 South Street, 14th Floor/Room 14471, Philadelphia, PA, 19146, USA.
| | - Joseph M Kindler
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| |
Collapse
|
23
|
Arponen M, Jalava N, Widjaja N, Ivaska KK. Glucose transporters GLUT1, GLUT3, and GLUT4 have different effects on osteoblast proliferation and metabolism. Front Physiol 2022; 13:1035516. [PMID: 36523556 PMCID: PMC9744933 DOI: 10.3389/fphys.2022.1035516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/15/2022] [Indexed: 03/05/2024] Open
Abstract
Bone is an active tissue that undergoes constant remodeling. Bone formation requires energy and one of the energy sources of bone-forming osteoblasts is glucose, which is transported inside the cells via glucose transporters. However, the role of class I glucose transporters in the differentiation and metabolism of osteoblasts and their precursors, bone marrow mesenchymal stromal cells (BMSCs) remains inconclusive. Our aim was to characterize the expression and contribution of main class I glucose transporters, GLUT1, GLUT3, and GLUT4, during osteoblast proliferation and differentiation. To investigate the role of each GLUT, we downregulated GLUTs with siRNA technology in primary rat BMSCs. Live-cell imaging and RNA-seq analysis was used to evaluate downstream pathways in silenced osteoblasts. Glucose transporters GLUT1, GLUT3, and GLUT4 had distinct expression patterns in osteoblasts. GLUT1 was abundant in BMSCs, but rapidly and significantly downregulated during osteoblast differentiation by up to 80% (p < 0.001). Similar downregulation was observed for GLUT4 (p < 0.001). In contrast, expression levels of GLUT3 remained stable during differentiation. Osteoblasts lacked GLUT2. Silencing of GLUT4 resulted in a significant decrease in proliferation and differentiation of preosteoblasts (p < 0.001) and several pathways related to carbohydrate metabolism and cell signaling were suppressed. However, silencing of GLUT3 resulted in increased proliferation (p < 0.001), despite suppression of several pathways involved in cellular metabolism, biosynthesis and actin organization. Silencing of GLUT1 had no effect on proliferation and less changes in the transcriptome. RNA-seq dataset further revealed that osteoblasts express also class II and III glucose transporters, except for GLUT7. In conclusion, GLUT1, -3 and -4 may all contribute to glucose uptake in differentiating osteoblasts. GLUT4 expression was clearly required for osteoblast proliferation and differentiation. GLUT1 appears to be abundant in early precursors, but stable expression of GLUT3 suggest also a role for GLUT3 in osteoblasts. Presence of other GLUT members may further contribute to fine-tuning of glucose uptake. Together, glucose uptake in osteoblast lineage appears to rely on several glucose transporters to ensure sufficient energy for new bone formation.
Collapse
Affiliation(s)
| | | | | | - Kaisa K. Ivaska
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
24
|
Zhu M, Fan Z. The role of the Wnt signalling pathway in the energy metabolism of bone remodelling. Cell Prolif 2022; 55:e13309. [PMID: 35811348 DOI: 10.1111/cpr.13309] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES Bone remodelling is necessary to repair old and impaired bone caused by aging and its effects. Injury in the process of bone remodelling generally leads to the development of various bone diseases. Energy metabolism plays crucial roles in bone cell formation and function, the disorder of which will disrupt the balance between bone formation and bone resorption. MATERIALS AND METHODS Here, we review the intrinsic interactions between bone remodelling and energy metabolism and the role of the Wnt signalling pathway. RESULTS We found a close interplay between metabolic pathways and bone homeostasis, demonstrating that bone plays an important role in the regulation of energy balance. We also discovered that Wnt signalling is associated with multiple biological processes regulating energy metabolism in bone cells. CONCLUSIONS Thus, targeted regulation of Wnt signalling and the recovery of the energy metabolism function of bone cells are key means for the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Mengyuan Zhu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Osteoblasts are responsible for bone matrix production during bone development and homeostasis. Much is known about the transcriptional regulation and signaling pathways governing osteoblast differentiation. However, less is known about how osteoblasts obtain or utilize nutrients to fulfill the energetic demands associated with osteoblast differentiation and bone matrix synthesis. The goal of this review is to highlight and discuss what is known about the role and regulation of bioenergetic metabolism in osteoblasts with a focus on more recent studies. RECENT FINDINGS Bioenergetic metabolism has emerged as an important regulatory node in osteoblasts. Recent studies have begun to identify the major nutrients and bioenergetic pathways favored by osteoblasts as well as their regulation during differentiation. Here, we highlight how osteoblasts obtain and metabolize glucose, amino acids, and fatty acids to provide energy and other metabolic intermediates. In addition, we highlight the signals that regulate nutrient uptake and metabolism and focus on how energetic metabolism promotes osteoblast differentiation. Bioenergetic metabolism provides energy and other metabolites that are critical for osteoblast differentiation and activity. This knowledge contributes to a more comprehensive understanding of osteoblast biology and may inform novel strategies to modulate osteoblast differentiation and bone anabolism in patients with bone disorders.
Collapse
Affiliation(s)
- Leyao Shen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guoli Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Courtney M Karner
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
26
|
Ye J, Xiao J, Wang J, Ma Y, Zhang Y, Zhang Q, Zhang Z, Yin H. The Interaction Between Intracellular Energy Metabolism and Signaling Pathways During Osteogenesis. Front Mol Biosci 2022; 8:807487. [PMID: 35155568 PMCID: PMC8832142 DOI: 10.3389/fmolb.2021.807487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/20/2021] [Indexed: 11/28/2022] Open
Abstract
Osteoblasts primarily mediate bone formation, maintain bone structure, and regulate bone mineralization, which plays an important role in bone remodeling. In the past decades, the roles of cytokines, signaling proteins, and transcription factors in osteoblasts have been widely studied. However, whether the energy metabolism of cells can be regulated by these factors to affect the differentiation and functioning of osteoblasts has not been explored in depth. In addition, the signaling and energy metabolism pathways are not independent but closely connected. Although energy metabolism is mediated by signaling pathways, some intermediates of energy metabolism can participate in protein post-translational modification. The content of intermediates, such as acetyl coenzyme A (acetyl CoA) and uridine diphosphate N-acetylglucosamine (UDP-N-acetylglucosamine), determines the degree of acetylation and glycosylation in terms of the availability of energy-producing substrates. The utilization of intracellular metabolic resources and cell survival, proliferation, and differentiation are all related to the integration of metabolic and signaling pathways. In this paper, the interaction between the energy metabolism pathway and osteogenic signaling pathway in osteoblasts and bone marrow mesenchymal stem cells (BMSCs) will be discussed.
Collapse
Affiliation(s)
- Jiapeng Ye
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Jirimutu Xiao
- Mongolian Medicine College, Inner Mongolia Medical University, Hohhot, China
| | - Jianwei Wang
- Department of Orthopedics and Traumatology, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- *Correspondence: Jianwei Wang, ; Heng Yin,
| | - Yong Ma
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Yafeng Zhang
- Department of Orthopedics and Traumatology, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Qiang Zhang
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Zongrui Zhang
- Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Heng Yin
- Department of Orthopedics and Traumatology, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- *Correspondence: Jianwei Wang, ; Heng Yin,
| |
Collapse
|
27
|
Zhou Y, Xu Z, Wang Y, Song Q, Yin R. LncRNA MALAT1 mediates osteogenic differentiation in osteoporosis by regulating the miR-485-5p/WNT7B axis. Front Endocrinol (Lausanne) 2022; 13:922560. [PMID: 36760811 PMCID: PMC9904362 DOI: 10.3389/fendo.2022.922560] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 12/28/2022] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION Accumulating evidence demonstrates that long non-coding RNAs (lncRNAs) are associated with the development of osteoporosis. METHODS This study aimed to investigate the effects of MALAT1 on osteogenic differentiation and cell apoptosis in osteoporosis. MALAT1 level, detected by RT-qPCR, was downregulated in hindlimb unloading (HU) mice and simulated microgravity (MG)-treated MC3T3-E1 cells. Moreover, osteogenic differentiation-related factor (Bmp4, Col1a1, and Spp1) levels were measured by RT-qPCR and Western blot. ALP activity was detected, and ALP staining was performed. Cell apoptosis was assessed by flow cytometry. RESULTS The results revealed that MALAT1 upregulated the expression of Bmp4, Col1a1, and Spp1, and enhanced ALP activity. Knockdown of MALAT1 suppressed their expression and ALP activity, suggesting that MALAT1 promoted osteogenic differentiation. Additionally, MALAT1 inhibited apoptosis, increased Bax and caspase-3 levels, and decreased Bcl-2 level. However, knockdown of MALAT1 had opposite results. In MG cells, MALAT1 facilitated osteogenic differentiation and suppressed apoptosis. Furthermore, miR-485-5p was identified as a target of MALAT1, and WNT7B was verified as a target of miR-485-5p. Overexpression of miR-485-5p rescued the promotion of osteogenic differentiation and the inhibition of apoptosis induced by MALAT1. Knockdown of WNT7B abolished the facilitation of osteogenic differentiation and the suppression of apoptosis induced by downregulation of miR-485-5p. DISCUSSION In conclusion, MALAT1 promoted osteogenic differentiation and inhibited cell apoptosis through the miR-485-5p/WNT7B axis, which suggested that MALAT1 is a potential target to alleviate osteoporosis.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Clinical Laboratory, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhuo Xu
- Department of Rehabilitation, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yuanyi Wang
- Department of Spine Surgery, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Qiang Song
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ruofeng Yin
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Ruofeng Yin,
| |
Collapse
|
28
|
Wu F, Li B, Hu X, Yu F, Shi Y, Ye L. Wnt7b Inhibits Osteoclastogenesis via AKT Activation and Glucose Metabolic Rewiring. Front Cell Dev Biol 2021; 9:771336. [PMID: 34881243 PMCID: PMC8645835 DOI: 10.3389/fcell.2021.771336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/12/2021] [Indexed: 02/05/2023] Open
Abstract
The imbalance between bone formation and bone resorption causes osteoporosis, which leads to severe bone fractures. It is known that increases in osteoclast numbers and activities are the main reasons for increasing bone resorption. Although extensive studies have investigated the regulation of osteoclastogenesis of bone marrow macrophages (BMMs), new pharmacological avenues still need to be unveiled for clinical purpose. Wnt ligands have been widely demonstrated as stimulators of bone formation; however, the inhibitory effect of the Wnt pathway in osteoclastogenesis is largely unknown. Here, we demonstrate that Wnt7b, a potent Wnt ligand that enhances bone formation and increases bone mass, also abolishes osteoclastogenesis in vitro. Importantly, enforced expression of Wnt in bone marrow macrophage lineage cells significantly disrupts osteoclast formation and activity, which leads to a dramatic increase in bone mass. Mechanistically, Wnt7b impacts the glucose metabolic process and AKT activation during osteoclastogenesis. Thus, we demonstrate that Wnt7b diminishes osteoclast formation, which will be beneficial for osteoporosis therapy in the future.
Collapse
Affiliation(s)
- Fanzi Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Boer Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuchen Hu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Lojk J, Marc J. Roles of Non-Canonical Wnt Signalling Pathways in Bone Biology. Int J Mol Sci 2021; 22:10840. [PMID: 34639180 PMCID: PMC8509327 DOI: 10.3390/ijms221910840] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/15/2023] Open
Abstract
The Wnt signalling pathway is one of the central signalling pathways in bone development, homeostasis and regulation of bone mineral density. It consists of numerous Wnt ligands, receptors and co-receptors, which ensure tight spatiotemporal regulation of Wnt signalling pathway activity and thus tight regulation of bone tissue homeostasis. This enables maintenance of optimal mineral density, tissue healing and adaptation to changes in bone loading. While the role of the canonical/β-catenin Wnt signalling pathway in bone homeostasis is relatively well researched, Wnt ligands can also activate several non-canonical, β-catenin independent signalling pathways with important effects on bone tissue. In this review, we will provide a thorough overview of the current knowledge on different non-canonical Wnt signalling pathways involved in bone biology, focusing especially on the pathways that affect bone cell differentiation, maturation and function, processes involved in bone tissue structure regulation. We will describe the role of the two most known non-canonical pathways (Wnt/planar cell polarity pathways and Wnt/Ca2+ pathway), as well as other signalling pathways with a strong role in bone biology that communicate with the Wnt signalling pathway through non-canonical Wnt signalling. Our goal is to bring additional attention to these still not well researched but important pathways in the regulation of bone biology in the hope of prompting additional research in the area of non-canonical Wnt signalling pathways.
Collapse
Affiliation(s)
- Jasna Lojk
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia;
- University Clinical Center Ljubljana, Clinical Department of Clinical Chemistry and Biochemistry, 1000 Ljubljana, Slovenia
| |
Collapse
|
30
|
Luo B, Zhou X, Tang Q, Yin Y, Feng G, Li S, Chen L. Circadian rhythms affect bone reconstruction by regulating bone energy metabolism. J Transl Med 2021; 19:410. [PMID: 34579752 PMCID: PMC8477514 DOI: 10.1186/s12967-021-03068-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/02/2021] [Indexed: 01/02/2023] Open
Abstract
Metabolism is one of the most complex cellular biochemical reactions, providing energy and substances for basic activities such as cell growth and proliferation. Early studies have shown that glucose is an important nutrient in osteoblasts. In addition, amino acid metabolism and fat metabolism also play important roles in bone reconstruction. Mammalian circadian clocks regulate the circadian cycles of various physiological functions. In vertebrates, circadian rhythms are mediated by a set of central clock genes: muscle and brain ARNT like-1 (Bmal1), muscle and brain ARNT like-2 (Bmal2), circadian rhythmic motion output cycle stagnates (Clock), cryptochrome 1 (Cry1), cryptochrome2 (Cry2), period 1 (Per1), period 2 (Per2), period 3 (Per3) and neuronal PAS domain protein 2 (Npas2). Negative feedback loops, controlled at both the transcriptional and posttranslational levels, adjust these clock genes in a diurnal manner. According to the results of studies on circadian transcriptomic studies in several tissues, most rhythmic genes are expressed in a tissue-specific manner and are affected by tissue-specific circadian rhythms. The circadian rhythm regulates several activities, including energy metabolism, feeding time, sleeping, and endocrine and immune functions. It has been reported that the circadian rhythms of mammals are closely related to bone metabolism. In this review, we discuss the regulation of the circadian rhythm/circadian clock gene in osteoblasts/osteoclasts and the energy metabolism of bone, and the relationship between circadian rhythm, bone remodeling, and energy metabolism. We also discuss the therapeutic potential of regulating circadian rhythms or changing energy metabolism on bone development/bone regeneration.
Collapse
Affiliation(s)
- Beibei Luo
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Xin Zhou
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Guangxia Feng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Shue Li
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
31
|
Jin Z, Kho J, Dawson B, Jiang MM, Chen Y, Ali S, Burrage LC, Grover M, Palmer DJ, Turner DL, Ng P, Nagamani SC, Lee B. Nitric oxide modulates bone anabolism through regulation of osteoblast glycolysis and differentiation. J Clin Invest 2021; 131:138935. [PMID: 33373331 DOI: 10.1172/jci138935] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 12/22/2020] [Indexed: 12/25/2022] Open
Abstract
Previous studies have shown that nitric oxide (NO) supplements may prevent bone loss and fractures in preclinical models of estrogen deficiency. However, the mechanisms by which NO modulates bone anabolism remain largely unclear. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing arginine, the sole precursor for nitric oxide synthase-dependent (NOS-dependent) NO synthesis. Moreover, ASL is also required for channeling extracellular arginine to NOS for NO production. ASL deficiency (ASLD) is thus a model to study cell-autonomous, NOS-dependent NO deficiency. Here, we report that loss of ASL led to decreased NO production and impairment of osteoblast differentiation. Mechanistically, the bone phenotype was at least in part driven by the loss of NO-mediated activation of the glycolysis pathway in osteoblasts that led to decreased osteoblast differentiation and function. Heterozygous deletion of caveolin 1, a negative regulator of NO synthesis, restored NO production, osteoblast differentiation, glycolysis, and bone mass in a hypomorphic mouse model of ASLD. The translational significance of these preclinical studies was further reiterated by studies conducted in induced pluripotent stem cells from an individual with ASLD. Taken together, our findings suggest that ASLD is a unique genetic model for studying NO-dependent osteoblast function and that the NO/glycolysis pathway may be a new target to modulate bone anabolism.
Collapse
Affiliation(s)
- Zixue Jin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Jordan Kho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Brian Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Saima Ali
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Monica Grover
- Department of Pediatric Endocrinology, Stanford School of Medicine, Stanford, California, USA
| | - Donna J Palmer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Dustin L Turner
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Philip Ng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Sandesh Cs Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
32
|
Kim S, Henneicke H, Cavanagh LL, Macfarlane E, Thai LJ, Foong D, Gasparini SJ, Fong-Yee C, Swarbrick MM, Seibel MJ, Zhou H. Osteoblastic glucocorticoid signaling exacerbates high-fat-diet- induced bone loss and obesity. Bone Res 2021; 9:40. [PMID: 34465731 PMCID: PMC8408138 DOI: 10.1038/s41413-021-00159-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/21/2021] [Accepted: 05/19/2021] [Indexed: 12/22/2022] Open
Abstract
Chronic high-fat diet (HFD) consumption not only promotes obesity and insulin resistance, but also causes bone loss through mechanisms that are not well understood. Here, we fed wild-type CD-1 mice either chow or a HFD (43% of energy from fat) for 18 weeks; HFD-fed mice exhibited decreased trabecular volume (-28%) and cortical thickness (-14%) compared to chow-fed mice. In HFD-fed mice, bone loss was due to reduced bone formation and mineral apposition, without obvious effects on bone resorption. HFD feeding also increased skeletal expression of sclerostin and caused deterioration of the osteocyte lacunocanalicular network (LCN). In mice fed HFD, skeletal glucocorticoid signaling was activated relative to chow-fed mice, independent of serum corticosterone concentrations. We therefore examined whether skeletal glucocorticoid signaling was necessary for HFD-induced bone loss, using transgenic mice lacking glucocorticoid signaling in osteoblasts and osteocytes (HSD2OB/OCY-tg mice). In HSD2OB/OCY-tg mice, bone formation and mineral apposition rates were not suppressed by HFD, and bone loss was significantly attenuated. Interestingly, in HSD2OB/OCY-tg mice fed HFD, both Wnt signaling (less sclerostin induction, increased β-catenin expression) and glucose uptake were significantly increased, relative to diet- and genotype-matched controls. The osteocyte LCN remained intact in HFD-fed HSD2OB/OCY-tg mice. When fed a HFD, HSD2OB/OCY-tg mice also increased their energy expenditure and were protected against obesity, insulin resistance, and dyslipidemia. Therefore, glucocorticoid signaling in osteoblasts and osteocytes contributes to the suppression of bone formation in HFD-fed mice. Skeletal glucocorticoid signaling is also an important determinant of glucose uptake in bone, which influences the whole-body metabolic response to HFD.
Collapse
Affiliation(s)
- Sarah Kim
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia.,Concord Clinical School, The University of Sydney, Sydney, NSW, Australia
| | - Holger Henneicke
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia.,Department of Medicine III, Technische University Dresden Medical Center, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany.,Center for Regenerative Therapies Dresden, Technische University Dresden, Dresden, Germany
| | - Lauryn L Cavanagh
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Eugenie Macfarlane
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Lee Joanne Thai
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Daphne Foong
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Sylvia J Gasparini
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Colette Fong-Yee
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Michael M Swarbrick
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia.,Concord Clinical School, The University of Sydney, Sydney, NSW, Australia
| | - Markus J Seibel
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia.,Concord Clinical School, The University of Sydney, Sydney, NSW, Australia.,Department of Endocrinology and Metabolism, Concord Repatriation General Hospital, The University of Sydney, Sydney, NSW, Australia
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW, Australia. .,Concord Clinical School, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
33
|
Chen H, Song F, Long F. WNT7B overexpression rescues bone loss caused by glucocorticoids in mice. FASEB J 2021; 35:e21683. [PMID: 34118078 PMCID: PMC12107615 DOI: 10.1096/fj.202100151rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 01/28/2023]
Abstract
Glucocorticoids, widely prescribed for anti-inflammatory and immunosuppressive purposes, are the most common secondary cause for osteoporosis and related fractures. Current anti-resorptive and anabolic therapies are insufficient for treating glucocorticoid-induced osteoporosis due to contraindications or concerns of side effects. Glucocorticoids have been shown to disrupt Wnt signaling in osteoblast-lineage cells, but the efficacy for Wnt proteins to restore bone mass after glucocorticoid therapy has not been examined. Here by using two mouse genetic models wherein WNT7B expression is temporally activated by either tamoxifen or doxycycline in osteoblast-lineage cells, we show that WNT7B recovers bone mass following glucocorticoid-induced bone loss, thanks to increased osteoblast number and function. However, WNT7B overexpression in bone either before or after glucocorticoid treatments does not ameliorate the abnormal accumulation of body fat. The study demonstrates a potent bone anabolic function for WNT7B in countering glucocorticoid-induced bone loss.
Collapse
Affiliation(s)
- Hong Chen
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangfang Song
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Fanxin Long
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
34
|
Loss of Wnt16 Leads to Skeletal Deformities and Downregulation of Bone Developmental Pathway in Zebrafish. Int J Mol Sci 2021; 22:ijms22136673. [PMID: 34206401 PMCID: PMC8268848 DOI: 10.3390/ijms22136673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022] Open
Abstract
Wingless-type MMTV integration site family, member 16 (wnt16), is a wnt ligand that participates in the regulation of vertebrate skeletal development. Studies have shown that wnt16 can regulate bone metabolism, but its molecular mechanism remains largely undefined. We obtained the wnt16−/− zebrafish model using the CRISPR-Cas9-mediated gene knockout screen with 11 bp deletion in wnt16, which led to the premature termination of amino acid translation and significantly reduced wnt16 expression, thus obtaining the wnt16−/− zebrafish model. The expression of wnt16 in bone-related parts was detected via in situ hybridization. The head, spine, and tail exhibited significant deformities, and the bone mineral density and trabecular bone decreased in wnt16−/− using light microscopy and micro-CT analysis. RNA sequencing was performed to explore the differentially expressed genes (DEGs). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis found that the down-regulated DEGs are mainly concentrated in mTOR, FoxO, and VEGF pathways. Protein–protein interaction (PPI) network analysis was performed with the detected DEGs. Eight down-regulated DEGs including akt1, bnip4, ptena, vegfaa, twsg1b, prkab1a, prkab1b, and pla2g4f.2 were validated by qRT-PCR and the results were consistent with the RNA-seq data. Overall, our work provides key insights into the influence of wnt16 gene on skeletal development.
Collapse
|
35
|
Abstract
The involvement of nitric oxide (NO) in preventing bone loss has long been hypothesized, but despite decades of research the mechanisms remain obscure. In this issue of the JCI, Jin et al. explored NO deficiency using human cell and mouse models that lacked argininosuccinate lyase (ASL), the enzyme involved in synthesizing arginine and NO production. Osteoblasts that did not express ASL produced less NO and failed to differentiate. Notably, in the context of Asl deficiency, heterozygous deletion of caveolin 1, which normally inhibits NO synthesis, restored NO production, osteoblast differentiation, glycolysis, and bone mass. These experiments suggest that ASL regulates arginine synthesis in osteoblasts, which leads to enhanced NO production and increased glucose metabolism. After a period when research slowed, these studies, like the legendary phoenix, renew the exploration of NO in bone biology, and provide exciting translational potential.
Collapse
Affiliation(s)
- Hanghang Liu
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | |
Collapse
|
36
|
Wu Z, Wen Y, Fan G, He H, Zhou S, Chen L. HEMGN and SLC2A1 might be potential diagnostic biomarkers of steroid-induced osteonecrosis of femoral head: study based on WGCNA and DEGs screening. BMC Musculoskelet Disord 2021; 22:85. [PMID: 33451334 PMCID: PMC7811219 DOI: 10.1186/s12891-021-03958-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/05/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Steroid-induced osteonecrosis of the femoral head (SONFH) is a chronic and crippling bone disease. This study aims to reveal novel diagnostic biomarkers of SONFH. METHODS The GSE123568 dataset based on peripheral blood samples from 10 healthy individuals and 30 SONFH patients was used for weighted gene co-expression network analysis (WGCNA) and differentially expressed genes (DEGs) screening. The genes in the module related to SONFH and the DEGs were extracted for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Genes with |gene significance| > 0.7 and |module membership| > 0.8 were selected as hub genes in modules. The DEGs with the degree of connectivity ≥5 were chosen as hub genes in DEGs. Subsequently, the overlapping genes of hub genes in modules and hub genes in DEGs were selected as key genes for SONFH. And then, the key genes were verified in another dataset, and the diagnostic value of key genes was evaluated by receiver operating characteristic (ROC) curve. RESULTS Nine gene co-expression modules were constructed via WGCNA. The brown module with 1258 genes was most significantly correlated with SONFH and was identified as the key module for SONFH. The results of functional enrichment analysis showed that the genes in the key module were mainly enriched in the inflammatory response, apoptotic process and osteoclast differentiation. A total of 91 genes were identified as hub genes in the key module. Besides, 145 DEGs were identified by DEGs screening and 26 genes were identified as hub genes of DEGs. Overlapping genes of hub genes in the key module and hub genes in DEGs, including RHAG, RNF14, HEMGN, and SLC2A1, were further selected as key genes for SONFH. The diagnostic value of these key genes for SONFH was confirmed by ROC curve. The validation results of these key genes in GSE26316 dataset showed that only HEMGN and SLC2A1 were downregulated in the SONFH group, suggesting that they were more likely to be diagnostic biomarkers of SOFNH than RHAG and RNF14. CONCLUSIONS Our study identified that two key genes, HEMGN and SLC2A1, might be potential diagnostic biomarkers of SONFH.
Collapse
Affiliation(s)
- Zhixin Wu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan City, 430071, Hubei Province, China
| | - Yinxian Wen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan City, 430071, Hubei Province, China.
| | - Guanlan Fan
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hangyuan He
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan City, 430071, Hubei Province, China
| | - Siqi Zhou
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan City, 430071, Hubei Province, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan City, 430071, Hubei Province, China.
| |
Collapse
|
37
|
van Gastel N, Carmeliet G. Metabolic regulation of skeletal cell fate and function in physiology and disease. Nat Metab 2021; 3:11-20. [PMID: 33398192 DOI: 10.1038/s42255-020-00321-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 01/28/2023]
Abstract
The skeleton is diverse in its functions, which include mechanical support, movement, blood cell production, mineral storage and endocrine regulation. This multifaceted role is achieved through an interplay of osteoblasts, chondrocytes, bone marrow adipocytes and stromal cells, all generated from skeletal stem cells. Emerging evidence shows the importance of cellular metabolism in the molecular control of the skeletal system. The different skeletal cell types not only have distinct metabolic demands relating to their particular functions but also are affected by microenvironmental constraints. Specific metabolites control skeletal stem cell maintenance, direct lineage allocation and mediate cellular communication. Here, we discuss recent findings on the roles of cellular metabolism in determining skeletal stem cell fate, coordinating osteoblast and chondrocyte function, and organizing stromal support of haematopoiesis. We also consider metabolic dysregulation in skeletal ageing and degenerative diseases, and provide an outlook on how the field may evolve in the coming years.
Collapse
Affiliation(s)
- Nick van Gastel
- de Duve Institute, Brussels, Belgium.
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium.
| |
Collapse
|
38
|
Role of Metabolism in Bone Development and Homeostasis. Int J Mol Sci 2020; 21:ijms21238992. [PMID: 33256181 PMCID: PMC7729585 DOI: 10.3390/ijms21238992] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Carbohydrates, fats, and proteins are the underlying energy sources for animals and are catabolized through specific biochemical cascades involving numerous enzymes. The catabolites and metabolites in these metabolic pathways are crucial for many cellular functions; therefore, an imbalance and/or dysregulation of these pathways causes cellular dysfunction, resulting in various metabolic diseases. Bone, a highly mineralized organ that serves as a skeleton of the body, undergoes continuous active turnover, which is required for the maintenance of healthy bony components through the deposition and resorption of bone matrix and minerals. This highly coordinated event is regulated throughout life by bone cells such as osteoblasts, osteoclasts, and osteocytes, and requires synchronized activities from different metabolic pathways. Here, we aim to provide a comprehensive review of the cellular metabolism involved in bone development and homeostasis, as revealed by mouse genetic studies.
Collapse
|
39
|
Lee WC, Ji X, Nissim I, Long F. Malic Enzyme Couples Mitochondria with Aerobic Glycolysis in Osteoblasts. Cell Rep 2020; 32:108108. [PMID: 32905773 PMCID: PMC8183612 DOI: 10.1016/j.celrep.2020.108108] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/24/2020] [Accepted: 08/13/2020] [Indexed: 01/12/2023] Open
Abstract
The metabolic program of osteoblasts, the chief bone-making cells, remains incompletely understood. Here in murine calvarial cells, we establish that osteoblast differentiation under aerobic conditions is coupled with a marked increase in glucose consumption and lactate production but reduced oxygen consumption. As a result, aerobic glycolysis accounts for approximately 80% of the ATP production in mature osteoblasts. In vivo tracing with 13C-labeled glucose in the mouse shows that glucose in bone is readily metabolized to lactate but not organic acids in the TCA cycle. Glucose tracing in osteoblast cultures reveals that pyruvate is carboxylated to form malate integral to the malate-aspartate shuttle. RNA sequencing (RNA-seq) identifies Me2, encoding the mitochondrial NAD-dependent isoform of malic enzyme, as being specifically upregulated during osteoblast differentiation. Knockdown of Me2 markedly reduces the glycolytic flux and impairs osteoblast proliferation and differentiation. Thus, the mitochondrial malic enzyme functionally couples the mitochondria with aerobic glycolysis in osteoblasts.
Collapse
Affiliation(s)
- Wen-Chih Lee
- Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, PA 19104, USA
| | - Xing Ji
- Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, PA 19104, USA
| | - Itzhak Nissim
- Division of Genetics and Metabolism, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopedics, The Children's Hospital of Philadelphia, PA 19104, USA; Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Yang J, Ueharu H, Mishina Y. Energy metabolism: A newly emerging target of BMP signaling in bone homeostasis. Bone 2020; 138:115467. [PMID: 32512164 PMCID: PMC7423769 DOI: 10.1016/j.bone.2020.115467] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022]
Abstract
Energy metabolism is the process of generating energy (i.e. ATP) from nutrients. This process is indispensable for cell homeostasis maintenance and responses to varying conditions. Cells require energy for growth and maintenance and have evolved to have multiple pathways to produce energy. Both genetic and functional studies have demonstrated that energy metabolism, such as glucose, fatty acid, and amino acid metabolism, plays important roles in the formation and function of bone cells including osteoblasts, osteocytes, and osteoclasts. Dysregulation of energy metabolism in bone cells consequently disturbs the balance between bone formation and bone resorption. Metabolic diseases have also been reported to affect bone homeostasis. Bone morphogenic protein (BMP) signaling plays critical roles in regulating the formation and function of bone cells, thus affecting bone development and homeostasis. Mutations of BMP signaling-related genes in mice have been reported to show abnormalities in energy metabolism in many tissues, including bone. In addition, BMP signaling correlates with critical signaling pathways such as mTOR, HIF, Wnt, and self-degradative process autophagy to coordinate energy metabolism and bone homeostasis. These findings will provide a newly emerging target of BMP signaling and potential therapeutic strategies and the improved management of bone diseases. This review summarizes the recent advances in our understanding of (1) energy metabolism in regulating the formation and function of bone cells, (2) function of BMP signaling in whole body energy metabolism, and (3) mechanistic interaction of BMP signaling with other signaling pathways and biological processes critical for energy metabolism and bone homeostasis.
Collapse
Affiliation(s)
- Jingwen Yang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| | - Hiroki Ueharu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
41
|
Luo Y, Ding X, Ji H, Li M, Song H, Li S, Wang C, Wu H, Du H. MicroRNA-503-3p affects osteogenic differentiation of human adipose-derived stem cells by regulation of Wnt2 and Wnt7b under cyclic strain. Stem Cell Res Ther 2020; 11:318. [PMID: 32711579 PMCID: PMC7382842 DOI: 10.1186/s13287-020-01842-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/05/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) play a role in regulating osteogenic differentiation (OD) of mesenchymal stem cells by inhibiting mRNAs translation under cyclic strain. miR-503-3p was downregulated in OD of human adipose-derived stem cells (hASCs) in vivo under cyclic strain in our previous study, while it might target the Wnt/β-catenin (W-β) pathway. In this study, we explored miR-503-3p's role in OD of hASCs under cyclic strain. METHODS OD of hASCs was induced by cyclic strain. Bioinformatic and dual luciferase analyses were used to confirm the relationship between Wnt2/Wnt7b and miR-503-3p. Immunofluorescence was used to detect the effect of miR-503-3p on Wnt2/Wnt7b and β-catenin in hASCs transfected with miR-503-3p mimic and inhibitor. Mimic, inhibitor, and small interfering RNA (siRNA) transfected in hASCs to against Wnt2 and Wnt7b. Quantitative real-time PCR (RT-PCR) and western blot were used to examine the OD and W-β pathway at the mRNA and protein levels, respectively. Immunofluorescence was performed to locate β-catenin. ALP activity and calcium were detected by colorimetric assay. RESULTS Results of immunophenotypes by flow cytometry and multi-lineage potential confirmed that the cultured cells were hASCs. Results of luciferase reporter assay indicated that miR-503-3p could regulate the expression levels of Wnt2 and Wnt7b by targeting their respective 3'-untranslated region (UTR). Under cyclic strain, gain- or loss-function of miR-503-3p studies by mimic and inhibitor revealed that decreasing expression of miR-503-3p could significantly bring about promotion of OD of hASCs, whereas increased expression of miR-503-3p inhibited OD. Furthermore, miR-503-3p high-expression reduced the activity of the W-β pathway, as indicated by lowering expression of Wnt2 and Wnt7b, inactive β-catenin in miR-503-3p-treated hASCs. By contrast, miR-503-3p inhibition activated the W-β pathway. CONCLUSIONS Collectively, our findings indicate that miR-503-3p is a negative factor in regulating W-β pathway by Wnt2 and Wnt7b, which inhibit the OD of hASCs under cyclic strain.
Collapse
Affiliation(s)
- Yadong Luo
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Xu Ding
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Huan Ji
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Meng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Haiyang Song
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Sheng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Chenxing Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Heming Wu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China.,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China
| | - Hongming Du
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Nanjing Medical University, Hanzhong Road No.136, Nanjing, 210029, Jiangsu Province, People's Republic of China. .,Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, People's Republic of China.
| |
Collapse
|
42
|
Li Y, Liu Z, Tang Y, Feng W, Zhao C, Liao J, Zhang C, Chen H, Ren Y, Dong S, Liu Y, Hu N, Huang W. Schnurri-3 regulates BMP9-induced osteogenic differentiation and angiogenesis of human amniotic mesenchymal stem cells through Runx2 and VEGF. Cell Death Dis 2020; 11:72. [PMID: 31996667 PMCID: PMC6989499 DOI: 10.1038/s41419-020-2279-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Human amniotic mesenchymal stem cells (hAMSCs) are multiple potent progenitor cells (MPCs) that can differentiate into different lineages (osteogenic, chondrogenic, and adipogenic cells) and have a favorable capacity for angiogenesis. Schnurri-3 (Shn3) is a large zinc finger protein related to Drosophila Shn, which is a critical mediator of postnatal bone formation. Bone morphogenetic protein 9 (BMP9), one of the most potent osteogenic BMPs, can strongly upregulate various osteogenesis- and angiogenesis-related factors in MSCs. It remains unclear how Shn3 is involved in BMP9-induced osteogenic differentiation coupled with angiogenesis in hAMSCs. In this investigation, we conducted a comprehensive study to identify the effect of Shn3 on BMP9-induced osteogenic differentiation and angiogenesis in hAMSCs and analyze the responsible signaling pathway. The results from in vitro and in vivo experimentation show that Shn3 notably inhibits BMP9-induced early and late osteogenic differentiation of hAMSCs, expression of osteogenesis-related factors, and subcutaneous ectopic bone formation from hAMSCs in nude mice. Shn3 also inhibited BMP9-induced angiogenic differentiation, expression of angiogenesis-related factors, and subcutaneous vascular invasion in mice. Mechanistically, we found that Shn3 prominently inhibited the expression of BMP9 and activation of the BMP/Smad and BMP/MAPK signaling pathways. In addition, we further found activity on runt-related transcription factor 2 (Runx2), vascular endothelial growth factor (VEGF), and the target genes shared by BMP and Shn3 signaling pathways. Silencing Shn3 could dramatically enhance the expression of Runx2, which directly regulates the downstream target VEGF to couple osteogenic differentiation with angiogenesis. To summarize, our findings suggested that Shn3 significantly inhibited the BMP9-induced osteogenic differentiation and angiogenesis in hAMSCs. The effect of Shn3 was primarily seen through inhibition of the BMP/Smad signaling pathway and depressed expression of Runx2, which directly regulates VEGF, which couples BMP9-induced osteogenic differentiation with angiogenesis.
Collapse
Affiliation(s)
- Yuwan Li
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ziming Liu
- Institute of Sports Medicine of China, Peking University Third Hospital, Beijing, 100191, China
| | - Yaping Tang
- Department of Stomatology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Wei Feng
- Laboratory of Skeletal Development and Regeneration, School of Life Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Chen Zhao
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junyi Liao
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengmin Zhang
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Hong Chen
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Youliang Ren
- Department of Orthopaedics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Shiwu Dong
- Department of Orthopaedics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yi Liu
- Department of Orthopaedics, the First Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Ning Hu
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Huang
- Department of Orthopaedics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
43
|
Abstract
The 11 existing FDA-approved osteoporosis drug treatments include hormone replacement therapy, 2 SERMs (raloxifene and bazedoxifene), 5 inhibitors of bone-resorbing osteoclasts (4 bisphosphonates and anti-RANKL denosumab), 2 parathyroid hormone analogues (teriparatide and abaloparatide), and 1 WNT signaling enhancer (romosozumab). These therapies are effective and provide multiple options for patients and physicians. As the genomic revolution continues, potential novel targets for future drug development are identified. This review takes a wide perspective to describe potentially rewarding topics to explore, including knowledge of genes and pathways involved in bone cell metabolism, the utility of animal models, targeting drugs to bone, and ongoing advances in drug design and delivery.
Collapse
|
44
|
Alekos NS, Moorer MC, Riddle RC. Dual Effects of Lipid Metabolism on Osteoblast Function. Front Endocrinol (Lausanne) 2020; 11:578194. [PMID: 33071983 PMCID: PMC7538543 DOI: 10.3389/fendo.2020.578194] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
The skeleton is a dynamic and metabolically active organ with the capacity to influence whole body metabolism. This newly recognized function has propagated interest in the connection between bone health and metabolic dysfunction. Osteoblasts, the specialized mesenchymal cells responsible for the production of bone matrix and mineralization, rely on multiple fuel sources. The utilization of glucose by osteoblasts has long been a focus of research, however, lipids and their derivatives, are increasingly recognized as a vital energy source. Osteoblasts possess the necessary receptors and catabolic enzymes for internalization and utilization of circulating lipids. Disruption of these processes can impair osteoblast function, resulting in skeletal deficits while simultaneously altering whole body lipid homeostasis. This article provides an overview of the metabolism of postprandial and stored lipids and the osteoblast's ability to acquire and utilize these molecules. We focus on the requirement for fatty acid oxidation and the pathways regulating this function as well as the negative impact of dyslipidemia on the osteoblast and skeletal health. These findings provide key insights into the nuances of lipid metabolism in influencing skeletal homeostasis which are critical to appreciate the extent of the osteoblast's role in metabolic homeostasis.
Collapse
Affiliation(s)
- Nathalie S. Alekos
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Megan C. Moorer
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Baltimore Veterans Administration Medical Center, Baltimore, MD, United States
| | - Ryan C. Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Baltimore Veterans Administration Medical Center, Baltimore, MD, United States
- *Correspondence: Ryan C. Riddle
| |
Collapse
|