1
|
Zhang B, He W, Pei Z, Guo Q, Wang J, Sun M, Yang X, Ariben J, Li S, Feng W, Meng C, Zhao Z, Sun C, Hu X, Bai R, Wang X, Hao T. Plasma proteins, circulating metabolites mediate causal inference studies on the effect of gut bacteria on the risk of osteoporosis development. Ageing Res Rev 2024; 101:102479. [PMID: 39214170 DOI: 10.1016/j.arr.2024.102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/29/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The role of gut bacteria in preventing and delaying osteoporosis has been studied. However, the causal relationship between gut bacteria, plasma proteins, circulating metabolites and osteoporosis (OP) risk has not been fully revealed. MATERIALS AND METHODS In this study, a two-sample Mendelian randomization study (MR) approach was used to assess the causal associations between gut bacteria, plasma proteins and circulating metabolites, and osteoporosis risk using Genome Wide Association Study (GWAS) data from gut bacteria(n=8208), plasma proteins(n=2263), circulating metabolites (n=123), and osteoporosis (3203 cases and 16380452 controls). Inverse-variance weighted (IVW) was used as the main analytical method to estimate the MR causal effect and to perform directional sensitivity analysis of causality. Finally, the mediating effect values for the influence of gut flora on OP pathogenesis through circulating metabolites were calculated by univariate MR analysis, and multivariate MR analysis. Next, we evaluated the effect of Phosphatidylcholine on the osteogenic function of bone marrow mesenchymal stem cells (BMSCs) through relevant experiments, including Edu detection of cell proliferation, alkaline phosphatase (ALP) staining, Alizarin red staining to evaluate osteogenic function, qPCR and WB detection of osteogenic differentiation related gene expression. RESULTS A total of 9 gut microbial taxa, 15 plasma proteins and eight circulating metabolites were analysed for significant causal associations with the development of OP. Significant causal effects of 7 on gut bacteria, plasma proteins and circulating metabolites were analysed by univariate MR analysis and these results were used as exposure factors for subsequent multivariate MR. Multivariate MR analyses yielded a significant effect of circulating metabolites Phosphatidylcholine and other cholines on OP (P<0.05). Further mediation effect analysis showed that the mediation effect of Bifidobacteriaceae affecting OP through the circulating metabolite Phosphatidylcholine and other cholines was -0.0224, with a 95 % confidence interval for the mediation effect that did not include 0, and the complete mediation effect was significant. Phosphatidylcholine can promote BMSCs proliferation and osteogenesis. CONCLUSION Our study demonstrated significant causal associations of gut bacteria, plasma proteins and circulating metabolites on OP, and that Bifidobacteriaceae affect OP through the circulating metabolites Phosphatidylcholine and other cholines. Phosphatidylcholine affects the osteogenic ability of BMSCs. Further exploration of potential microbiota-associated mechanisms of bone metabolism may offer new avenues for osteoporosis prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Baoxin Zhang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China; Tianjin Hospital, Tianjin University, Tianjin 300211, China.
| | - Wanxiong He
- Inner Mongolia Medical University, Hohhot 010050, China.
| | - Zhiwei Pei
- Tianjin Hospital, Tianjin University, Tianjin 300211, China.
| | - Qingwen Guo
- Key Laboratory of Biological Manufacturing in Inner Mongolia Autonomous Region, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China.
| | - Jianzhong Wang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Mingqi Sun
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Xiaolong Yang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | | | - Siqin Li
- Bayannur City Hospital, Bayannaoer 015000, China
| | - Wei Feng
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Chenyang Meng
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Zhenqun Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Chao Sun
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Xiaoxin Hu
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Rui Bai
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
| | - Xing Wang
- Bayannur City Hospital, Bayannaoer 015000, China.
| | - Ting Hao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.
| |
Collapse
|
2
|
Zhu J, Li J, Yao T, Li T, Chang B, Yi X. Analysis of the role of irisin receptor signaling in regulating osteogenic/adipogenic differentiation of bone marrow mesenchymal stem cells. Biotechnol Genet Eng Rev 2024; 40:2012-2035. [PMID: 37010292 DOI: 10.1080/02648725.2023.2197713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/27/2023] [Indexed: 04/04/2023]
Abstract
The study aimed to explore the role of the irisin receptor (integrin αVβ5) signaling pathway in obesity-induced osteoporosis and its potential mechanism. The integrin αVβ5 gene of bone marrow mesenchymal stem cells (BMSCs) was silenced and overexpressed, and the cells were exposed to irisin treatment and mechanical stretch. Mouse models of obesity were established by feeding mice a high-fat diet, and 8-week caloric restriction/aerobic exercise regimens were implemented. The results showed that after silencing the integrin αVβ5, the osteogenic differentiation of BMSCs was significantly reduced. While overexpression of the integrin αVβ5 increased the osteogenic differentiation of BMSCs. Besides, mechanical stretch promoted the osteogenic differentiation of BMSCs. Obesity did not affect integrin αVβ5 expression in the bone, but it downregulated the expression of irisin and osteogenic factors, upregulated the expression of adipogenic factors, increased bone marrow fat, reduced bone formation, and destroyed the bone microstructure. Caloric restriction, exercise, and a combined regimen reversed these effects and improved obesity-induced osteoporosis, with the combined treatment exhibiting the most potent effect. This study confirms that the irisin receptor signaling pathway has a significant part in transmitting 'mechanical stress' and regulating 'osteogenic/adipogenic differentiation' of BMSCs via recombinant irisin, mechanical stretch, and overexpression/silencing of the integrin αVβ5 gene.
Collapse
Affiliation(s)
- Jingsheng Zhu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Department of Physical Education, Zhengzhou University, Zhengzhou, China
| | - Jing Li
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Tingting Yao
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Tao Li
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Bo Chang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Xuejie Yi
- School of Kinesiology, Shenyang Sport University, Shenyang, China
| |
Collapse
|
3
|
Birks S, Howard S, O’Rourke C, Thompson WR, Lau A, Uzer G. Osterix-driven LINC complex disruption in vivo diminishes osteogenesis at 8 weeks but not at 15 weeks. J Orthop Res 2024; 42:2007-2016. [PMID: 38602438 PMCID: PMC11293982 DOI: 10.1002/jor.25849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/12/2024]
Abstract
The Linker of Nucleoskeleton and Cytoskeleton (LINC) complex is a crucial connective component between the nuclear envelope and the cytoskeleton involving various cellular processes including nuclear positioning, nuclear architecture, and mechanotransduction. How LINC complexes regulate bone formation in vivo, however, is not well understood. To start bridging this gap, here we created a LINC disruption murine model using transgenic mice expressing Cre recombinase enzyme under the control of the Osterix (Osx-Cre) which is primarily active in pre-osteoblasts and floxed Tg(CAG-LacZ/EGFP-KASH2) mice. Tg(CAG-LacZ/EGFP-KASH2) mice contain a lox-STOP-lox flanked LacZ gene which is deleted upon cre recombination allowing for the overexpression of an EGFP-KASH2 fusion protein. This overexpressed protein disrupts endogenous Nesprin-Sun binding leading to disruption of LINC complexes. Thus, crossing these two lines results in an Osx- driven LINC disruption (ODLD) specific to pre-osteoblasts. In this study, we investigated how this LINC disruption affects exercise-induced bone accrual. ODLD cells had decreased osteogenic and adipogenic potential in vitro compared to non-disrupted controls and sedentary ODLD mice showed decreased bone quality at 8 weeks. Upon access to a voluntary running wheel, ODLD animals showed increased running time and distance; however, our 6-week exercise intervention did not significantly affect bone microarchitecture and bone mechanical properties.
Collapse
Affiliation(s)
- Scott Birks
- Boise State University, Micron School of Materials Science and Engineering
| | - Sean Howard
- Boise State University, Mechanical and Biomedical Engineering
| | | | | | - Anthony Lau
- The College of New Jersey, Biomedical Engineering
| | - Gunes Uzer
- Boise State University, Mechanical and Biomedical Engineering
| |
Collapse
|
4
|
Wang W, Qian J, Shang M, Qiao Y, Huang J, Gao X, Ye Z, Tong X, Xu K, Li X, Liu Z, Zhou L, Zheng S. Integrative analysis of the transcriptome and metabolome reveals the importance of hepatokine FGF21 in liver aging. Genes Dis 2024; 11:101161. [PMID: 39022127 PMCID: PMC11252782 DOI: 10.1016/j.gendis.2023.101161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 07/20/2024] Open
Abstract
Aging is a contributor to liver disease. Hence, the concept of liver aging has become prominent and has attracted considerable interest, but its underlying mechanism remains poorly understood. In our study, the internal mechanism of liver aging was explored via multi-omics analysis and molecular experiments to support future targeted therapy. An aged rat liver model was established with d-galactose, and two other senescent hepatocyte models were established by treating HepG2 cells with d-galactose and H2O2. We then performed transcriptomic and metabolomic assays of the aged liver model and transcriptome analyses of the senescent hepatocyte models. In livers, genes related to peroxisomes, fatty acid elongation, and fatty acid degradation exhibited down-regulated expression with aging, and the hepatokine Fgf21 expression was positively correlated with the down-regulation of these genes. In senescent hepatocytes, similar to the results found in aged livers, FGF21 expression was also decreased. Moreover, the expressions of cell cycle-related genes were significantly down-regulated, and the down-regulated gene E2F8 was the key cell cycle-regulating transcription factor. We then validated that FGF21 overexpression can protect against liver aging and that FGF21 can attenuate the declines in the antioxidant and regenerative capacities in the aging liver. We successfully validated the results from cellular and animal experiments using human liver and blood samples. Our study indicated that FGF21 is an important target for inhibiting liver aging and suggested that pharmacological prevention of the reduction in FGF21 expression due to aging may be used to treat liver aging-related diseases.
Collapse
Affiliation(s)
- Wenchao Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Junjie Qian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Mingge Shang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Jiacheng Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Xinxin Gao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Zhou Ye
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Xinyu Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Kangdi Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Xiang Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Zhengtao Liu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang 310015, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310000, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, Zhejiang 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou, Zhejiang 310003, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
5
|
Ojala R, Widjaja N, Hentilä J, Jalo A, Helin JS, Nissinen TA, Jalava N, Eskola O, Rajander J, Löyttyniemi E, Ivaska KK, Hannukainen JC. Evaluation of bone marrow glucose uptake and adiposity in male rats after diet and exercise interventions. Front Endocrinol (Lausanne) 2024; 15:1422869. [PMID: 38948514 PMCID: PMC11211282 DOI: 10.3389/fendo.2024.1422869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024] Open
Abstract
Objectives Obesity impairs bone marrow (BM) glucose metabolism. Adult BM constitutes mostly of adipocytes that respond to changes in energy metabolism by modulating their morphology and number. Here we evaluated whether diet or exercise intervention could improve the high-fat diet (HFD) associated impairment in BM glucose uptake (BMGU) and whether this associates with the morphology of BM adipocytes (BMAds) in rats. Methods Eight-week-old male Sprague-Dawley rats were fed ad libitum either HFD or chow diet for 24 weeks. Additionally after 12 weeks, HFD-fed rats switched either to chow diet, voluntary intermittent running exercise, or both for another 12 weeks. BMAd morphology was assessed by perilipin-1 immunofluorescence staining in formalin-fixed paraffin-embedded tibial sections. Insulin-stimulated sternal and humeral BMGU were measured using [18F]FDG-PET/CT. Tibial microarchitecture and mineral density were measured with microCT. Results HFD rats had significantly higher whole-body fat percentage compared to the chow group (17% vs 13%, respectively; p = 0.004) and larger median size of BMAds in the proximal tibia (815 µm2 vs 592 µm2, respectively; p = 0.03) but not in the distal tibia. Switch to chow diet combined with running exercise normalized whole-body fat percentage (p < 0.001) but not the BMAd size. At 32 weeks of age, there was no significant difference in insulin-stimulated BMGU between the study groups. However, BMGU was significantly higher in sternum compared to humerus (p < 0.001) and higher in 8-week-old compared to 32-week-old rats (p < 0.001). BMAd size in proximal tibia correlated positively with whole-body fat percentage (r = 0.48, p = 0.005) and negatively with humeral BMGU (r = -0.63, p = 0.02). HFD significantly reduced trabecular number (p < 0.001) compared to the chow group. Switch to chow diet reversed this as the trabecular number was significantly higher (p = 0.008) than in the HFD group. Conclusion In this study we showed that insulin-stimulated BMGU is age- and site-dependent. BMGU was not affected by the study interventions. HFD increased whole-body fat percentage and the size of BMAds in proximal tibia. Switching from HFD to a chow diet and running exercise improved glucose homeostasis and normalized the HFD-induced increase in body fat but not the hypertrophy of BMAds.
Collapse
Affiliation(s)
- Ronja Ojala
- Turku PET Centre, University of Turku, Turku, Finland
| | - Nicko Widjaja
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Anna Jalo
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Jatta S. Helin
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Tuuli A. Nissinen
- MediCity Research Laboratory, University of Turku, Turku, Finland
- Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Niki Jalava
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Olli Eskola
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Johan Rajander
- Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, University of Turku, Turku, Finland
- Department of Biostatistics, Turku University Hospital, Turku, Finland
| | - Kaisa K. Ivaska
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | |
Collapse
|
6
|
Alonso N, Almer G, Semeraro MD, Rodriguez-Blanco G, Fauler G, Anders I, Ritter G, vom Scheidt A, Hammer N, Gruber HJ, Herrmann M. Impact of High-Fat Diet and Exercise on Bone and Bile Acid Metabolism in Rats. Nutrients 2024; 16:1744. [PMID: 38892677 PMCID: PMC11174439 DOI: 10.3390/nu16111744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Bile acids help facilitate intestinal lipid absorption and have endocrine activity in glucose, lipid and bone metabolism. Obesity and exercise influence bile acid metabolism and have opposite effects in bone. This study investigates if regular exercise helps mitigate the adverse effects of obesity on bone, potentially by reversing alterations in bile acid metabolism. Four-month-old female Sprague Dawley rats either received a high-fat diet (HFD) or a chow-based standard diet (lean controls). During the 10-month study period, half of the animals performed 30 min of running at moderate speed on five consecutive days followed by two days of rest. The other half was kept inactive (inactive controls). At the study's end, bone quality was assessed by microcomputed tomography and biomechanical testing. Bile acids were measured in serum and stool. HFD feeding was related to reduced trabecular (-33%, p = 1.14 × 10-7) and cortical (-21%, p = 2.9 × 10-8) bone mass and lowered femoral stiffness (12-41%, p = 0.005). Furthermore, the HFD decreased total bile acids in serum (-37%, p = 1.0 × 10-6) but increased bile acids in stool (+2-fold, p = 7.3 × 10-9). These quantitative effects were accompanied by changes in the relative abundance of individual bile acids. The concentration of serum bile acids correlated positively with all cortical bone parameters (r = 0.593-0.708), whilst stool levels showed inverse correlations at the cortical (r = -0.651--0.805) and trabecular level (r = -0.656--0.750). Exercise improved some trabecular and cortical bone quality parameters (+11-31%, p = 0.043 to 0.001) in lean controls but failed to revert the bone loss related to the HFD. Similarly, changes in bile acid metabolism were not mitigated by exercise. Prolonged HFD consumption induced quantitative and qualitative alterations in bile acid metabolism, accompanied by bone loss. Tight correlations between bile acids and structural indices of bone quality support further functional analyses on the potential role of bile acids in bone metabolism. Regular moderate exercise improved trabecular and cortical bone quality in lean controls but failed in mitigating the effects related to the HFD in bone and bile acid metabolism.
Collapse
Affiliation(s)
- Nerea Alonso
- Clinical Institute for Medical and Chemical Laboratory Diagnostics (CIMCL), Medical University of Graz, 8036 Graz, Austria
| | - Gunter Almer
- Clinical Institute for Medical and Chemical Laboratory Diagnostics (CIMCL), Medical University of Graz, 8036 Graz, Austria
| | - Maria Donatella Semeraro
- Clinical Institute for Medical and Chemical Laboratory Diagnostics (CIMCL), Medical University of Graz, 8036 Graz, Austria
| | - Giovanny Rodriguez-Blanco
- Clinical Institute for Medical and Chemical Laboratory Diagnostics (CIMCL), Medical University of Graz, 8036 Graz, Austria
- LKH-Universitätsklinikum Graz, 8036 Graz, Austria
| | - Günter Fauler
- Clinical Institute for Medical and Chemical Laboratory Diagnostics (CIMCL), Medical University of Graz, 8036 Graz, Austria
| | - Ines Anders
- Division of Biomedical Research, Medical University of Graz, 8036 Graz, Austria (G.R.)
| | - Gerald Ritter
- Division of Biomedical Research, Medical University of Graz, 8036 Graz, Austria (G.R.)
| | | | - Niels Hammer
- Department of Anatomy, Medical University of Graz, 8036 Graz, Austria
- Department of Orthopaedic and Trauma Surgery, University of Leipzig, 04103 Leipzig, Germany
- Division of Biomechatronics, Fraunhofer Institute for Machine Tools and Forming Technology, 01187 Dresden, Germany
| | - Hans-Jürgen Gruber
- Clinical Institute for Medical and Chemical Laboratory Diagnostics (CIMCL), Medical University of Graz, 8036 Graz, Austria
| | - Markus Herrmann
- Clinical Institute for Medical and Chemical Laboratory Diagnostics (CIMCL), Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
7
|
Rutkowsky JM, Wong A, Toupadakis CA, Rutledge JC, Yellowley CE. Lipolysis products from triglyceride-rich lipoproteins induce stress protein ATF3 in osteoblasts. J Orthop Res 2024; 42:1033-1044. [PMID: 38044472 PMCID: PMC11009083 DOI: 10.1002/jor.25756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/07/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
High fat diets overwhelm the physiological mechanisms for absorption, storage, and utilization of triglycerides (TG); consequently TG, TG-rich lipoproteins (TGRL), and TGRL remnants accumulate, circulate systemically, producing dyslipidemia. This associates with, or is causative for increased atherosclerotic cardiovascular risk, ischemic stroke, fatty liver disease, and pancreatitis. TGRL hydrolysis by endothelial surface-bound lipoprotein lipase (LPL) generates metabolites like free fatty acids which have proinflammatory properties. While osteoblasts utilize fatty acids as an energy source, dyslipidemia is associated with negative effects on the skeleton. In this study we investigated the effects of TGRL lipolysis products (TGRL-LP) on expression of a stress responsive transcription factor, termed activating transcription factor 3 (ATF3), reactive oxygen species (ROS), ATF3 target genes, and angiopoietin-like 4 (Angptl4) in osteoblasts. As ATF3 negatively associates with osteoblast differentiation, we also investigated the skeletal effects of global ATF3 deletion in mice. TGRL-LP increased expression of Atf3, proinflammatory proteins Ptgs2 and IL-6, and induced ROS in MC3T3-E1 osteoblastic cells. Angptl4 is an endogenous inhibitor of LPL which was transcriptionally induced by TGRL-LP, while recombinant Angptl4 prevented TG-driven Atf3 induction. Atf3 global knockout male mice demonstrated increased trabecular and cortical microarchitectural parameters. In summary, we find that TGRL-LP induce osteoblastic cell stress as evidenced by expression of ATF3, which may contribute to the negative impact of dyslipidemia in the skeleton. Further, concomitant induction of Angptl4 in osteoblasts might play a protective role by reducing local lipolysis.
Collapse
Affiliation(s)
| | - Alice Wong
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine
| | | | - John C. Rutledge
- Department of Internal Medicine (Cardiology), School of Medicine, University of California Davis, Davis, CA 95616
| | - Clare E. Yellowley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine
| |
Collapse
|
8
|
Birks S, Howard S, Wright CS, O’Rourke C, Day EA, Lamb AJ, Walsdorf JR, Lau A, Thompson WR, Uzer G. Prrx1-driven LINC complex disruption in vivo reduces osteoid deposition but not bone quality after voluntary wheel running. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.22.559054. [PMID: 37790521 PMCID: PMC10542150 DOI: 10.1101/2023.09.22.559054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
The Linker of Nucleoskeleton and Cytoskeleton (LINC) complex serves to connect the nuclear envelope and the cytoskeleton, influencing cellular processes such as nuclear arrangement, architecture, and mechanotransduction. The role LINC plays in mechanotransduction pathways in bone progenitor cells has been well studied; however, the mechanisms by which LINC complexes govern in vivo bone formation remain less clear. To bridge this knowledge gap, we established a murine model disrupting LINC using transgenic Prx-Cre mice and floxed Tg(CAG-LacZ/EGFP-KASH2) mice. Prx-Cre mice express the Cre recombinase enzyme controlled by the paired-related homeobox gene-1 promoter (Prrx1), a pivotal regulator of skeletal development. Prx-Cre animals have been widely used in the bone field to target bone progenitor cells. Tg(CAG-LacZ/EGFP-KASH2) mice carry a lox-stop-lox flanked LacZ gene allowing for the overexpression of an EGFP-KASH2 fusion protein via cre recombinase mediated deletion of the LacZ cassette. This disrupts endogenous Nesprin-Sun binding in a dominant negative manner disconnecting nesprin from the nuclear envelope. By combining these lines, we generated a Prrx1(+) cell-specific LINC disruption model to study its impact on the developing skeleton and subsequently exercise-induced bone accrual. The findings presented here indicate Prx-driven LINC disruption (PDLD) cells exhibit no change in osteogenic and adipogenic potential compared to controls in vitro nor are there bone quality changes when compared to in sedentary animals at 8 weeks. While PDLD animals displayed increased voluntary running activity andPrrx1(+) cell-specific LINC disruption abolished the exercise-induced increases in osteoid volume and surface after a 6-week exercise intervention, no other changes in bone microarchitecture or mechanical properties were found.
Collapse
Affiliation(s)
- Scott Birks
- Boise State University, Micron School of Materials Science and Engineering
| | - Sean Howard
- Boise State University, Mechanical and Biomedical Engineering
| | - Christian S. Wright
- Indiana University, Department of Physical Therapy, School of Health and Human Sciences
| | | | - Elicza A. Day
- Indiana University, Department of Physical Therapy, School of Health and Human Sciences
| | - Alexander J. Lamb
- Indiana University, Department of Physical Therapy, School of Health and Human Sciences
| | - James R. Walsdorf
- Indiana University, Department of Physical Therapy, School of Health and Human Sciences
| | - Anthony Lau
- The College of New Jersey, Biomedical Engineering
| | - William R. Thompson
- Indiana University, Department of Physical Therapy, School of Health and Human Sciences
| | - Gunes Uzer
- Boise State University, Mechanical and Biomedical Engineering
| |
Collapse
|
9
|
Saito MK, de Oliveira BK, Macedo AP, Sorrentino Dos Santos C, Lopes RT, Yamanaka JS, Shimano AC. Cafeteria Diet Can Affect Bone Microarchitecture in Sedentary and Trained Male Rats. J Clin Densitom 2024; 27:101467. [PMID: 38306807 DOI: 10.1016/j.jocd.2024.101467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 12/31/2023] [Accepted: 01/18/2024] [Indexed: 02/04/2024]
Abstract
INTRODUCTION Poor eating habits and a sedentary lifestyle can impair health. Regular physical activity improves the quality of life and is essential for bone health. Therefore, the present study aimed to evaluate the effects of the cafeteria diet on bone quality of sedentary and exercised rats. METHODS Sixty young male Wistar rats were divided into six groups (n=10) according to diet composition and activity level, being: SD+CON, standard diet and control; SD+SED, standard diet and sedentary; SD+EX, standard diet and exercised; CD+CON, cafeteria diet and control; CD+SED, cafeteria diet and sedentary; CD+EX, cafeteria diet and exercised. The exercise protocol consisted of 10 ladder-climbing sessions/day, 5 days/week, and the sedentary rats were maintained in individual cages with limited mobility. Body mass and food intake were evaluated weekly. After 10 weeks, the animals were euthanized, and white adipose tissue was collected. The bone structure was evaluated by densitometry, mechanical tests, histomorphometric, and micro-computed tomography analyses. RESULTS The cafeteria diet increased adipose tissue (p<0.001), decreased bone mineral density (p=0.004), and impaired biomechanical properties (p<0.05) and histomorphometry parameters (p=0.044). The sedentarism decreased bone mineral density (p<0.001) and biomechanical properties (p<0.05), and the exercise did not improve bone properties. CONCLUSION In this experimental model, it was concluded that the cafeteria diet and a sedentary lifestyle negatively affect bone, and ladder-climbing exercise could not prevent the effects of the unhealthy diet.
Collapse
Affiliation(s)
- Marcio Koiti Saito
- Department of Orthopedics and Anesthesiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| | - Beatriz Kawano de Oliveira
- Department of Orthopedics and Anesthesiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Ana Paula Macedo
- Department of Dental Materials and Prosthesis, Faculty of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Ricardo Tadeu Lopes
- Nuclear Instrumentation Laboratory, COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jéssica Suzuki Yamanaka
- Department of Orthopedics and Anesthesiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Antonio Carlos Shimano
- Department of Orthopedics and Anesthesiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
10
|
Chargo NJ, Kang HJ, Das S, Jin Y, Rockwell C, Cho JY, McCabe LR, Parameswaran N. Korean red ginseng extract prevents bone loss in an oral model of glucocorticoid induced osteoporosis in mice. Front Pharmacol 2024; 15:1268134. [PMID: 38533264 PMCID: PMC10963623 DOI: 10.3389/fphar.2024.1268134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024] Open
Abstract
The gut microbiota and barrier function play important roles in bone health. We previously demonstrated that chronic glucocorticoid (GC)-induced bone loss in mice is associated with significant shifts in gut microbiota composition and impaired gut barrier function. Korean Red Ginseng (KRG, Panax Ginseng Meyer, Araliaceae) extract has been shown to prevent glucocorticoid-induced osteoporosis (GIO) in a subcutaneous pellet model in mice, but its effect on gut microbiota and barrier function in this context is not known. The overall goal of this study was to test the effect of KRG extract in a clinically relevant, oral model of GIO and further investigate its role in modulating the gut-bone axis. Growing male mice (CD-1, 8 weeks) were treated with 75 μg/mL corticosterone (∼9 mg/kg/day) or 0.4% ethanol vehicle in the drinking water for 4 weeks. During this 4-week period, mice were treated daily with 500 mg/kg/day KRG extract dissolved in sterile water or an equal amount of sterile water via oral gastric gavage. After 4 weeks of treatment, we assessed bone volume, microbiota composition, gut barrier integrity, and immune cells in the bone marrow (BM) and mesenteric lymph nodes (MLNs). 4 weeks of oral GC treatment caused significant distal femur trabecular bone loss, and this was associated with changes in gut microbiota composition, impaired gut barrier function and altered immune cell composition. Importantly, KRG extract prevented distal femur trabecular bone loss and caused significant alterations in gut microbiota composition but had only modest effects on gut barrier function and immune cell populations. Taken together, these results demonstrate that KRG extract significantly modulates the gut microbiota-bone axis and prevents glucocorticoid-induced bone loss in mice.
Collapse
Affiliation(s)
- Nicholas J. Chargo
- Department of Physiology, Michigan State University, East Lansing, MI, United States
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| | - Ho Jun Kang
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Subhashari Das
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Yining Jin
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Cheryl Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Laura R. McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, United States
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, MI, United States
- College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
11
|
Zhang YW, Wu Y, Liu XF, Chen X, Su JC. Targeting the gut microbiota-related metabolites for osteoporosis: The inextricable connection of gut-bone axis. Ageing Res Rev 2024; 94:102196. [PMID: 38218463 DOI: 10.1016/j.arr.2024.102196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Osteoporosis is a systemic skeletal disease characterized by decreased bone mass, destruction of bone microstructure, raised bone fragility, and enhanced risk of fractures. The correlation between gut microbiota and bone metabolism has gradually become a widespread research hotspot in recent years, and successive studies have revealed that the alterations of gut microbiota and its-related metabolites are related to the occurrence and progression of osteoporosis. Moreover, several emerging studies on the relationship between gut microbiota-related metabolites and bone metabolism are also underway, and extensive research evidence has indicated an inseparable connection between them. Combined with latest literatures and based on inextricable connection of gut-bone axis, this review is aimed to summarize the relation, potential mechanisms, application strategies, clinical application prospects, and existing challenges of gut microbiota and its-related metabolites on osteoporosis, thus updating the knowledge in this research field and providing certain reference for future researches.
Collapse
Affiliation(s)
- Yuan-Wei Zhang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Xiang-Fei Liu
- Department of Orthopaedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Xiao Chen
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai 200092, China.
| | - Jia-Can Su
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
12
|
Zhang YW, Song PR, Wang SC, Liu H, Shi ZM, Su JC. Diets intervene osteoporosis via gut-bone axis. Gut Microbes 2024; 16:2295432. [PMID: 38174650 PMCID: PMC10773645 DOI: 10.1080/19490976.2023.2295432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Osteoporosis is a systemic skeletal disease that seriously endangers the health of middle-aged and older adults. Recently, with the continuous deepening of research, an increasing number of studies have revealed gut microbiota as a potential target for osteoporosis, and the research concept of the gut-bone axis has gradually emerged. Additionally, the intake of dietary nutrients and the adoption of dietary patterns may affect the gut microbiota, and alterations in the gut microbiota might also influence the metabolic status of the host, thus adjusting bone metabolism. Based on the gut-bone axis, dietary intake can also participate in the modulation of bone metabolism by altering abundance, diversity, and composition of gut microbiota. Herein, combined with emerging literatures and relevant studies, this review is aimed to summarize the impacts of different dietary components and patterns on osteoporosis by acting on gut microbiota, as well as underlying mechanisms and proper dietary recommendations.
Collapse
Affiliation(s)
- Yuan-Wei Zhang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Pei-Ran Song
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Si-Cheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Zhong-Min Shi
- Department of Orthopaedics, Sixth People’s Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jia-Can Su
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Organoid Research Center, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
13
|
Li Z, Wang Q, Huang X, Wu Y, Shan D. Microbiome's role in musculoskeletal health through the gut-bone axis insights. Gut Microbes 2024; 16:2410478. [PMID: 39387683 PMCID: PMC11469435 DOI: 10.1080/19490976.2024.2410478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/06/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
The interplay between the human microbiome and the musculoskeletal system represents a burgeoning field of research with profound implications for understanding and treating musculoskeletal disorders. This review articulates the pivotal role of the microbiome in modulating bone health, highlighting the gut-bone axis as a critical nexus for potential therapeutic intervention. Through a meticulous analysis of recent clinical research, we underscore the microbiome's influence on osteoporosis, sarcopenia, osteoarthritis, and rheumatoid arthritis, delineating both the direct and indirect mechanisms by which microbiota could impact musculoskeletal integrity and function. Our investigation reveals novel insights into the microbiota's contribution to bone density regulation, hormone production, immune modulation, and nutrient absorption, laying the groundwork for innovative microbiome-based strategies in musculoskeletal disease management. Significantly, we identify the challenges hindering the translation of research into clinical practice, including the limitations of current microbial sequencing techniques and the need for standardized methodologies in microbiome studies. Furthermore, we highlight promising directions for future research, particularly in the realm of personalized medicine, where the microbiome's variability offers unique opportunities for tailored treatment approaches. This review sets a new agenda for leveraging gut microbiota in the diagnosis, prevention, and treatment of musculoskeletal conditions, marking a pivotal step toward integrating microbiome science into clinical musculoskeletal care.
Collapse
Affiliation(s)
- Zhengrui Li
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Wang
- Jiangsu University, Zhenjiang, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Yinteng Wu
- Department of Orthopedic and Trauma Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Dan Shan
- Faculty of Health and Medicine, Lancaster University, Lancaster, UK
- Department of Biobehavioral Sciences, Columbia University, New York, NY, USA
| |
Collapse
|
14
|
Wang W, Xu K, Shang M, Li X, Tong X, Liu Z, Zhou L, Zheng S. The biological mechanism and emerging therapeutic interventions of liver aging. Int J Biol Sci 2024; 20:280-295. [PMID: 38164175 PMCID: PMC10750291 DOI: 10.7150/ijbs.87679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/22/2023] [Indexed: 01/03/2024] Open
Abstract
Research on liver aging has become prominent and has attracted considerable interest in uncovering the mechanism and therapeutic targets of aging to expand lifespan. In addition, multi-omics studies are widely used to perform further mechanistic investigations on liver aging. In this review, we illustrate the changes that occur with aging in the liver, present the current models of liver aging, and emphasize existing multi-omics studies on liver aging. We integrated the multi-omics data of enrolled studies and reanalyzed them to identify key pathways and targets of liver aging. The results indicated that C-X-C motif chemokine ligand 9 (Cxcl9) was a regulator of liver aging. In addition, we provide a flowchart for liver aging research using multi-omics analysis and molecular experiments to help researchers conduct further research. Finally, we present emerging therapeutic treatments that prolong lifespan. In summary, using cells and animal models of liver aging, we can apply a multi-omics approach to find key metabolic pathways and target genes to mitigate the adverse effects of liver aging.
Collapse
Affiliation(s)
- Wenchao Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Kangdi Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Mingge Shang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Xiang Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Xinyu Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Zhengtao Liu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou 310000, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China
- Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou 310000, China
| |
Collapse
|
15
|
Birks S, Howard S, O’Rourke C, Thompson WR, Lau A, Uzer G. Osterix-driven LINC complex disruption in vivo diminishes bone microarchitecture in 8-week male mice but not after 6-week voluntary wheel running. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554623. [PMID: 37662368 PMCID: PMC10473717 DOI: 10.1101/2023.08.24.554623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The Linker of Nucleoskeleton and Cytoskeleton (LINC) complex is a crucial connective component between the nuclear envelope and the cytoskeleton involving various cellular processes including nuclear positioning, nuclear architecture, and mechanotransduction. How LINC complexes regulate bone formation in vivo, however, is not well understood. To start bridging this gap, here we created a LINC disruption murine model using transgenic mice expressing Cre recombinase enzyme under the control of the Osterix (Osx-Cre) which is primarily active in pre-osteoblasts and floxed Tg(CAG-LacZ/EGFP-KASH2) mice. Tg(CAG-LacZ/EGFP-KASH2) mice contain a lox-STOP-lox flanked LacZ gene which is deleted upon cre recombination allowing for the overexpression of an EGFP-KASH2 fusion protein. This overexpressed protein disrupts endogenous Nesprin-Sun binding leading to disruption of LINC complexes. Thus, crossing these two lines results in a Osx-driven LINC disruption (ODLD) specific to pre-osteoblasts. In this study, we investigated how this LINC disruption affects exercise induced bone accrual. ODLD cells had decreased osteogenic and adipogenic potential in vitro compared to non-disrupted controls and sedentary ODLD mice showed decreased bone quality at 8-weeks. Upon access to a voluntary running wheel ODLD animals showed increased running time and distance; however, our 6-week exercise intervention did not significantly affect bone microarchitecture and bone mechanical properties.
Collapse
Affiliation(s)
- Scott Birks
- Boise State University, Micron School of Materials Science and Engineering
| | - Sean Howard
- Boise State University, Mechanical and Biomedical Engineering
| | | | | | - Anthony Lau
- The College of New Jersey, Biomedical Engineering
| | - Gunes Uzer
- Boise State University, Mechanical and Biomedical Engineering
| |
Collapse
|
16
|
Raji-Amirhasani A, Khaksari M, Soltani Z, Saberi S, Iranpour M, Darvishzadeh Mahani F, Hajializadeh Z, Sabet N. Beneficial effects of time and energy restriction diets on the development of experimental acute kidney injury in Rat: Bax/Bcl-2 and histopathological evaluation. BMC Nephrol 2023; 24:59. [PMID: 36941590 PMCID: PMC10026443 DOI: 10.1186/s12882-023-03104-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/03/2023] [Indexed: 03/23/2023] Open
Abstract
People's lifestyles and, especially, their eating habits affect their health and the functioning of the organs in their bodies, including the kidneys. One's diet influences the cells' responses to stressful conditions such as acute kidney injury (AKI). This study aims to determine the preconditioning effects of four different diets: energy restriction (ER) diet, time restriction (TR) eating, intermittent fasting (IF), and high-fat diet (HF) on histopathological indices of the kidney as well as the molecules involved in apoptosis during AKI. Adult male rats underwent ER, TR, IF, and HF diets for eight weeks. Then, AKI was induced, and renal function indices, histopathological indices, and molecules involved in apoptosis were measured. In animals with AKI, urinary albumin excretion, serum urea, creatinine and, Bax/Bcl-2 ratio increased in the kidney, while renal eGFR decreased. ER and TR diets improved renal parameters and prevented an increase in the Bax/Bcl-2 ratio. The IF diet improved renal parameters but had no effect on the Bax/Bcl-2 ratio. On the other hand, the HF diet worsened renal function and increased the Bax/Bcl-2 ratio. Histopathological examination also showed improved kidney conditions in the ER and TR groups and more damage in the HF group. This study demonstrated that ER and TR diets have renoprotective effects on AKI and possibly cause the resistance of kidney cells to damage by reducing the Bax/Bcl-2 ratio and improving apoptotic conditions.
Collapse
Affiliation(s)
- Alireza Raji-Amirhasani
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran.
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Shadan Saberi
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Iranpour
- Pathology and Stem Cells Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pathology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Darvishzadeh Mahani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Cardiovascular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Hajializadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Cardiovascular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Nazanin Sabet
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
17
|
Nota MH, Nicolas S, O’Leary OF, Nolan YM. Outrunning a bad diet: interactions between exercise and a Western-style diet for adolescent mental health, metabolism and microbes. Neurosci Biobehav Rev 2023; 149:105147. [PMID: 36990371 DOI: 10.1016/j.neubiorev.2023.105147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Adolescence is a period of biological, psychological and social changes, and the peak time for the emergence of mental health problems. During this life stage, brain plasticity including hippocampal neurogenesis is increased, which is crucial for cognitive functions and regulation of emotional responses. The hippocampus is especially susceptible to environmental and lifestyle influences, mediated by changes in physiological systems, resulting in enhanced brain plasticity but also an elevated risk for developing mental health problems. Indeed, adolescence is accompanied by increased activation of the maturing hypothalamic-pituitary-adrenal axis, sensitivity to metabolic changes due to increased nutritional needs and hormonal changes, and gut microbiota maturation. Importantly, dietary habits and levels of physical activity significantly impact these systems. In this review, the interactions between exercise and Western-style diets, which are high in fat and sugar, on adolescent stress susceptibility, metabolism and the gut microbiota are explored. We provide an overview of current knowledge on implications of these interactions for hippocampal function and adolescent mental health, and speculate on potential mechanisms which require further investigation.
Collapse
|
18
|
Liu T, Yu H, Wang S, Li H, Du X, He X. Chondroitin sulfate alleviates osteoporosis caused by calcium deficiency by regulating lipid metabolism. Nutr Metab (Lond) 2023; 20:6. [PMID: 36747190 PMCID: PMC9901125 DOI: 10.1186/s12986-023-00726-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/21/2023] [Indexed: 02/08/2023] Open
Abstract
The use of non-drug intervention for calcium deficiency has attracted attention in recent years. Although calcium carbonate is the preferred raw material for calcium supplementation, there are few reports on the mechanism of the combined action of chondroitin sulfate and calcium to alleviate osteoporosis from the perspective of gut microbiota and metabolomics. In this study, a rat model of osteoporosis was established by feeding a low-calcium diet. The intestinal microbiota abundance, fecal and plasma metabolite expression levels of rats fed a basal diet, a low-calcium diet, a low-calcium diet plus calcium carbonate, and a low-calcium diet plus chondroitin sulfate were compared. The results showed that compared with the low calcium group, the calcium content and bone mineral density of femur were significantly increased in the calcium carbonate and chondroitin sulfate groups. 16 S rRNA sequencing and metabolomics analysis showed that chondroitin sulfate intervention could reduce short-chain fatty acid synthesis of intestinal flora, slow down inflammatory response, inhibit osteoclast differentiation, promote calcium absorption and antioxidant mechanism, and alleviate osteoporosis in low-calcium feeding rats. Correlation analysis showed that the selected intestinal flora was significantly correlated with metabolites enriched in feces and plasma. This study provides scientific evidence of the potential impact of chondroitin sulfate as a dietary supplement for patients with osteoporosis.
Collapse
Affiliation(s)
- Tianshu Liu
- grid.27255.370000 0004 1761 1174Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012 Shandong China ,grid.27255.370000 0004 1761 1174Institute for Medical Dataology, Shandong University, National Institute of Health Data Science of China, Jinan, 250012 Shandong China
| | - Hai Yu
- grid.272242.30000 0001 2168 5385Division of Cancer RNA Research, National Cancer Center Research Institute, Tokyo, 104-0045 Japan ,grid.27255.370000 0004 1761 1174Institute for Medical Dataology, Shandong University, National Institute of Health Data Science of China, Jinan, 250012 Shandong China
| | - Shuai Wang
- grid.27255.370000 0004 1761 1174Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jining, 250012 Shandong China
| | - Huimin Li
- grid.27255.370000 0004 1761 1174Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012 Shandong China ,grid.506261.60000 0001 0706 7839National Human Genetic Resources Center; National Research Institute for Health and Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China
| | - Xinyiran Du
- grid.449428.70000 0004 1797 7280College of Stomatology, Jining Medical University, Jining, 272067 Shandong China
| | - Xiaodong He
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Institute for Medical Dataology, Shandong University, National Institute of Health Data Science of China, Jinan, 250012, Shandong, China.
| |
Collapse
|
19
|
Cellular and Molecular Mechanisms Associating Obesity to Bone Loss. Cells 2023; 12:cells12040521. [PMID: 36831188 PMCID: PMC9954309 DOI: 10.3390/cells12040521] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Obesity is an alarming disease that favors the upset of other illnesses and enhances mortality. It is spreading fast worldwide may affect more than 1 billion people by 2030. The imbalance between excessive food ingestion and less energy expenditure leads to pathological adipose tissue expansion, characterized by increased production of proinflammatory mediators with harmful interferences in the whole organism. Bone tissue is one of those target tissues in obesity. Bone is a mineralized connective tissue that is constantly renewed to maintain its mechanical properties. Osteoblasts are responsible for extracellular matrix synthesis, while osteoclasts resorb damaged bone, and the osteocytes have a regulatory role in this process, releasing growth factors and other proteins. A balanced activity among these actors is necessary for healthy bone remodeling. In obesity, several mechanisms may trigger incorrect remodeling, increasing bone resorption to the detriment of bone formation rates. Thus, excessive weight gain may represent higher bone fragility and fracture risk. This review highlights recent insights on the central mechanisms related to obesity-associated abnormal bone. Publications from the last ten years have shown that the main molecular mechanisms associated with obesity and bone loss involve: proinflammatory adipokines and osteokines production, oxidative stress, non-coding RNA interference, insulin resistance, and changes in gut microbiota. The data collection unveils new targets for prevention and putative therapeutic tools against unbalancing bone metabolism during obesity.
Collapse
|
20
|
Bermudez B, Ishii T, Wu YH, Carpenter RD, Sherk VD. Energy Balance and Bone Health: a Nutrient Availability Perspective. Curr Osteoporos Rep 2023; 21:77-84. [PMID: 36542294 DOI: 10.1007/s11914-022-00765-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Obesity is highly prevalent and is associated with bone fragility and fracture. The changing nutrient availability to bone in obesity is an important facet of bone health. The goal of this article is to summarize current knowledge on the effects of carbohydrate and dietary fat availability on bone, particularly in the context of other tissues. RECENT FINDINGS The skeleton is a primary site for fatty acid and glucose uptake. The trafficking of carbohydrates and fats into tissues changes with weight loss and periods of weight gain. Exercise acutely influences nutrient uptake into bone and may affect nutrient partitioning to bone. Bone cells secrete hormones that signal to the brain and other tissues information about its energetic state, which may alter whole-body nutrient trafficking. There is a critical need for studies to address the changes that metabolic perturbations have on nutrient availability in bone.
Collapse
Affiliation(s)
- Beatriz Bermudez
- Department of Mechanical Engineering, University of Colorado Denver, Denver, CO, USA
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Toru Ishii
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yuan-Haw Wu
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - R Dana Carpenter
- Department of Mechanical Engineering, University of Colorado Denver, Denver, CO, USA
| | - Vanessa D Sherk
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Division of Translational and Clinical Sciences, Center for Scientific Review, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Chlebek C, Rosen CJ. The Role of Bone Cell Energetics in Altering Bone Quality and Strength in Health and Disease. Curr Osteoporos Rep 2023; 21:1-10. [PMID: 36435911 DOI: 10.1007/s11914-022-00763-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Bone quality and strength are diminished with age and disease but can be improved by clinical intervention. Energetic pathways are essential for cellular function and drive osteogenic signaling within bone cells. Altered bone quality is associated with changes in the energetic activity of bone cells following diet-based or therapeutic interventions. Energetic pathways may directly or indirectly contribute to changes in bone quality. The goal of this review is to highlight tissue-level and bioenergetic changes in bone health and disease. RECENT FINDINGS Bone cell energetics are an expanding field of research. Early literature primarily focused on defining energetic activation throughout the lifespan of bone cells. Recent studies have begun to connect bone energetic activity to health and disease. In this review, we highlight bone cell energetic demands, the effect of substrate availability on bone quality, altered bioenergetics associated with disease treatment and development, and additional biological factors influencing bone cell energetics. Bone cells use several energetic pathways during differentiation and maturity. The orchestration of bioenergetic pathways is critical for healthy cell function. Systemic changes in substrate availability alter bone quality, potentially due to the direct effects of altered bone cell bioenergetic activity. Bone cell bioenergetics may also contribute directly to the development and treatment of skeletal diseases. Understanding the role of energetic pathways in the cellular response to disease will improve patient treatment.
Collapse
Affiliation(s)
- Carolyn Chlebek
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, USA.
| |
Collapse
|
22
|
Shi Z, Wang L, Luan J, Yin L, Ji X, Zhang W, Xu B, Chen L, He Y, Wang R, Liu L. Exercise Promotes Bone Marrow Microenvironment by Inhibiting Adipsin in Diet-Induced Male Obese Mice. Nutrients 2022; 15:nu15010019. [PMID: 36615677 PMCID: PMC9823335 DOI: 10.3390/nu15010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Obesity is a growing global epidemic linked to many diseases, including diabetes, cardiovascular diseases, and musculoskeletal disorders. Exercise can improve bone density and decrease excess bone marrow adipose tissue (BMAT) in obese individuals. However, the mechanism of exercise regulating bone marrow microenvironment remains unclear. This study examines how exercise induces bone marrow remodeling in diet-induced obesity. We employed unbiased RNA-Seq to investigate the effect of exercise on the bone marrow of diet-induced obese male mice. Bone mesenchymal stem cells (BMSCs) were isolated to explore the regulatory effects of exercise in vitro. Our data demonstrated that exercise could slow down the progression of obesity and improve trabecular bone density. RNA-seq data revealed that exercise inhibited secreted phosphoprotein 1 (Spp1), which was shown to mediate bone resorption through mechanosensing mechanisms. Interactome analysis of Spp1 using the HINT database showed that Spp1 interacted with the adipokine adipsin. Moreover, exercise decreased BMAT, which induced osteoclast differentiation and promoted bone loss. Our study reveals that exercise improves the bone marrow microenvironment by at least partially inhibiting the adipsin-Spp1 signaling pathway so as to inhibit the alternative complement system from activating osteoclasts in diet-induced obese mice.
Collapse
Affiliation(s)
- Zunhan Shi
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Lihui Wang
- Department of Medical Imaging, Shanghai East Hospital (East Hospital Affiliated to Tongji University), Tongji University, Shanghai 200123, China
| | - Jinwen Luan
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Liqin Yin
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaohui Ji
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Wenqian Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Bingxiang Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Linshan Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Ying He
- Department of Pathology and Cellular Biology and Naomi Berrie Diabetes Center, Columbia University, New York, NY 10027, USA
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
- Correspondence: (R.W.); (L.L.)
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
- Correspondence: (R.W.); (L.L.)
| |
Collapse
|
23
|
Rubin J, Styner M. The skeleton in a physical world. Exp Biol Med (Maywood) 2022; 247:2213-2222. [PMID: 35983849 PMCID: PMC9899984 DOI: 10.1177/15353702221113861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
All organisms exist within a physical space and respond to physical forces as part of daily life. In higher organisms, the skeleton is critical for locomotion in the physical environment, providing a carapace upon which the animal can move to accomplish functions necessary for living. As such, the skeleton has responded evolutionarily, and does in real-time, to physical stresses placed on it to ensure that its structure supports its function in the sea, in the air, and on dry land. In this article, we consider how those cells responsible for remodeling skeletal structure respond to mechanical force including load magnitude, frequency, and cyclicity, and how force rearranges cellular structure in turn. The effects of these forces to balance the mesenchymal stem cell supply of bone-forming osteoblasts and energy storing adipocytes are addressed. That this phenotypic switching is achieved at the level of both gene transactivation and alteration of structural epigenetic controls of gene expression is considered. Finally, as clinicians, we consider this information as it applies to a prescriptive for intelligent exercise.
Collapse
|
24
|
Zhang YW, Cao MM, Li YJ, Chen XX, Yu Q, Rui YF. A narrative review of the moderating effects and repercussion of exercise intervention on osteoporosis: ingenious involvement of gut microbiota and its metabolites. J Transl Med 2022; 20:490. [PMID: 36303163 DOI: 10.1186/s12967-022-03700-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/03/2022] [Accepted: 10/09/2022] [Indexed: 11/10/2022] Open
Abstract
Osteoporosis (OP) is a systemic bone disease characterized by the decreased bone mass and destruction of bone microstructure, which tends to result in the enhanced bone fragility and related fractures, as well as high disability rate and mortality. Exercise is one of the most common, reliable and cost-effective interventions for the prevention and treatment of OP currently, and numerous studies have revealed the close association between gut microbiota (GM) and bone metabolism recently. Moreover, exercise can alter the structure, composition and abundance of GM, and further influence the body health via GM and its metabolites, and the changes of GM also depend on the choice of exercise modes. Herein, combined with relevant studies and based on the inseparable relationship between exercise intervention-GM-OP, this review is aimed to discuss the moderating effects and potential mechanisms of exercise intervention on GM and bone metabolism, as well as the interaction between them.
Collapse
Affiliation(s)
- Yuan-Wei Zhang
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, 210009, Nanjing, Jiangsu, PR China.,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda Hospital, Southeast University, Nanjing Jiangsu, PR China.,School of Medicine, Southeast University, Nanjing, Jiangsu, PR China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
| | - Mu-Min Cao
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, 210009, Nanjing, Jiangsu, PR China.,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda Hospital, Southeast University, Nanjing Jiangsu, PR China.,School of Medicine, Southeast University, Nanjing, Jiangsu, PR China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
| | - Ying-Juan Li
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda Hospital, Southeast University, Nanjing Jiangsu, PR China.,Department of Geriatrics, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, PR China
| | - Xiang-Xu Chen
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, 210009, Nanjing, Jiangsu, PR China.,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda Hospital, Southeast University, Nanjing Jiangsu, PR China.,School of Medicine, Southeast University, Nanjing, Jiangsu, PR China.,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China
| | - Qian Yu
- Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda Hospital, Southeast University, Nanjing Jiangsu, PR China.,Department of Gastroenterology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, PR China
| | - Yun-Feng Rui
- Department of Orthopaedics, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Ding Jia Qiao, 210009, Nanjing, Jiangsu, PR China. .,Multidisciplinary Team (MDT) for Geriatric Hip Fracture Management, School of Medicine, Zhongda Hospital, Southeast University, Nanjing Jiangsu, PR China. .,School of Medicine, Southeast University, Nanjing, Jiangsu, PR China. .,Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
25
|
Zhang Z, Zhang Z, Pei L, Zhang X, Li B, Meng Y, Zhou X. How high-fat diet affects bone in mice: A systematic review and meta-analysis. Obes Rev 2022; 23:e13493. [PMID: 35822276 DOI: 10.1111/obr.13493] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/18/2022] [Accepted: 06/19/2022] [Indexed: 11/29/2022]
Abstract
High-fat diet (HFD) feeding for mice is commonly used to model obesity. However, conflicting results have been reported on the relationship between HFD and bone mass. In this systematic review and meta-analysis, we synthesized data from 80 articles to determine the alterations in cortical and trabecular bone mass of femur, tibia, and vertebrae in C57BL/6 mice after HFD. Overall, we detected decreased trabecular bone mass as well as deteriorated architecture, in femur and tibia of HFD treated mice. The vertebral trabecula was also impaired, possibly due to its reshaping into a more fragmentized pattern. In addition, pooled cortical thickness declined in femur, tibia, and vertebrae. Combined with changes in other cortical parameters, HFD could lead to a larger femoral bone marrow cavity, and a thinner and more fragile cortex. Moreover, we conducted subgroup analyses to explore the influence of mice's sex and age as well as HFD's ingredients and intervention period. Based on our data, male mice or mice aged 6-12 weeks old are relatively susceptible to HFD. HFD with > 50% of energy from fats and intervention time of 10 weeks to 5 months are more likely to induce skeletal alterations. Altogether, these findings supported HFD as an appropriate model for obesity-associated bone loss and can guide future studies.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, People's Republic of China
| | - Zhanrong Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, People's Republic of China
| | - Lei Pei
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaozhou Zhang
- College of Letters & Science, University of California Berkeley, Berkeley, California, USA
| | - Boyuan Li
- Fountain Valley School of Colorado, Colorado Springs, Colorado, USA
| | - Yichen Meng
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, People's Republic of China
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, People's Republic of China
| |
Collapse
|
26
|
Yan Q, Cai L, Guo W. New Advances in Improving Bone Health Based on Specific Gut Microbiota. Front Cell Infect Microbiol 2022; 12:821429. [PMID: 35860378 PMCID: PMC9289272 DOI: 10.3389/fcimb.2022.821429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 06/01/2022] [Indexed: 12/31/2022] Open
Abstract
The gut microbiota has been shown to play an important role in the pathogenesis of various diseases, including metabolic diseases, cardiovascular diseases, and cancer. Recent studies suggest that the gut microbiota is also closely associated with bone metabolism. However, given the high diversity of the gut microbiota, the effects of different taxa and compositions on bone are poorly understood. Previous studies demonstrated that the mechanisms underlying the effects of the gut microbiota on bone mainly include its modulation of nutrient absorption, intestinal permeability, metabolites (such as short-chain amino acids), immune responses, and hormones or neurotransmitters (such as 5-hydroxytryptamine). Several studies found that external interventions, such as dietary changes, improved bone health and altered the composition of the gut microbiota. This review summarises the beneficial gut bacteria and explores how dietary, natural, and physical factors alter the diversity and composition of the gut microbiota to improve bone health, thereby providing potential new insight into the prevention of osteoporosis.
Collapse
|
27
|
Sun X, Huang Y, Zhu S, Yan J, Gan K, Xu Z, Wang S, Kang X, Zhang J, Sun W. Yishen Qingli Heluo Granule in the Treatment of Chronic Kidney Disease: Network Pharmacology Analysis and Experimental Validation. Drug Des Devel Ther 2022; 16:769-787. [PMID: 35355655 PMCID: PMC8959874 DOI: 10.2147/dddt.s348335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/09/2022] [Indexed: 12/16/2022] Open
Abstract
Background Chronic kidney disease (CKD) is considered a global public health problem with high morbidity and mortality. Yishen Qingli Heluo granule (YQHG) is representative traditional Chinese medicine (TCM) remedy for clinical treatment of CKD. This study aims to explore the mechanism of YQHG on CKD through network pharmacology and experimental validation. Methods Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and wide-scale literature mining were applied to screen active compounds of YQHG. Multiple bioinformatic tools and online databases were applied by us to obtain relevant targets of YQHG and CKD. The intersection targets between YQHG and CKD were considered as candidate targets. The compound-target, herb-candidate target and protein–protein interaction networks were constructed and visualized for topological analyses. GO and KEGG enrichment analyses were conducted to determine the biological processes and signaling pathways. Molecular docking was used to verify the reliability of network pharmacology. Finally, pharmacological evaluation was performed to explore the mechanism of YQHG against CKD on a 5/6 nephrectomy model. Results Seventy-nine candidate targets, ten core biological processes and one key signaling pathway (p53) were screened. PTGS2 was identified as a key target based on H-CT network. The molecular docking showed that Quercetin, Kaempferol, Luteolin were three key compounds with the best binding activity. In addition, IL6 and Quercetin could form a stable complex with high binding affinity (−7.29 kcal/mol). In vivo experiment revealed that YQHG improved kidney function and fibrosis in 5/6 nephrectomized rats. Moreover, the decreased expression of PTGS2, IL6, and the increased expression of p53 were observed in kidney tissue. Notably, the gut microbiota of rats treated with YQHG was reshaped, which was characterized by a reduced ratio of Firmicutes/Bacteroidota. Conclusion Our results predicted and verified the potential targets of YQHG on CKD from a holistic perspective, and provided valuable direction for the further research of YQHG. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/02pumepfbPI
Collapse
Affiliation(s)
- Xian Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yiting Huang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Sha Zhu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Jin Yan
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Ke Gan
- Department of Rheumatology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| | - Zijing Xu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Shuaishuai Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Xiaoyu Kang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Junfeng Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Wei Sun
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People's Republic of China
| |
Collapse
|
28
|
Orwoll ES, Parimi N, Wiedrick J, Lapidus J, Napoli N, Wilkinson JE, Huttenhower C, Langsetmo L, Kiel DP. Analysis of the Associations Between the Human Fecal Microbiome and Bone Density, Structure, and Strength: The Osteoporotic Fractures in Men (MrOS) Cohort. J Bone Miner Res 2022; 37:597-607. [PMID: 35119137 PMCID: PMC9605688 DOI: 10.1002/jbmr.4518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/27/2021] [Accepted: 01/13/2022] [Indexed: 11/08/2022]
Abstract
In preclinical models, the composition and function of the gut microbiota have been linked to bone growth and homeostasis, but there are few available data from studies of human populations. In a hypothesis-generating experiment in a large cohort of community-dwelling older men (n = 831; age range, 78-98 years), we explored the associations between fecal microbial profiles and bone density, microarchitecture, and strength measured with total hip dual-energy X-ray absorptiometry (DXA) and high-resolution peripheral quantitative computed tomography (HRpQCT) (distal radius, distal and diaphyseal tibia). Fecal samples were collected and the 16S rRNA gene V4 hypervariable region sequenced. Sequences were bioinformatically processed through the DADA2 pipeline and then taxonomically assigned using SILVA. Generalized linear models as implemented in microbiome multivariable association with linear models (MaAsLin 2) were used to test for associations between skeletal measures and specific microbial genera. The abundances of four bacterial genera were weakly associated with bone density, structure, or strength (false discovery rate [FDR] ≤ 0.05), and the measured directions of associations of genera were generally consistent across multiple bone measures, supporting a role for microbiota on skeletal homeostasis. However, the associated effect sizes were small (log2 fold change < ±0.35), limiting power to confidently identify these associations even with high resolution skeletal imaging phenotypes, and we assessed the resulting implications for the design of future cohort-based studies. As in analogous examples from genomewide association studies, we find that larger cohort sizes will likely be needed to confidently identify associations between the fecal microbiota and skeletal health relying on 16S sequencing. Our findings bolster the view that the gut microbiome is associated with clinically important measures of bone health, while also indicating the challenges in the design of cohort-based microbiome studies. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Eric S Orwoll
- Department of Medicine, Oregon Health & Sciences University, Portland, OR, USA
| | - Neeta Parimi
- San Francisco Coordinating Center, San Francisco, CA, USA
| | - Jack Wiedrick
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
| | - Jodi Lapidus
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA.,Oregon Health & Science University - Portland State University School of Public Health, Portland, OR, USA
| | - Nicola Napoli
- Department of Medicine, Unit of Endocrinology and Diabetes, Campus Bio-Medico University of Rome, Rome, Italy.,Division of Bone and Mineral Diseases, Washington University, St Louis, MO, USA
| | - Jeremy E Wilkinson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lisa Langsetmo
- School of Public Health, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Douglas P Kiel
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew Senior Life, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
29
|
Wakefield CB, Lee VR, Johnston D, Boroumand P, Pillon NJ, Sayedyahossein S, O'Donnell BL, Tang J, Sanchez-Pupo RE, Barr KJ, Gros R, Flynn L, Borradaile NM, Klip A, Beier F, Penuela S. Pannexin 3 deletion reduces fat accumulation and inflammation in a sex-specific manner. Int J Obes (Lond) 2022; 46:726-738. [PMID: 34897286 DOI: 10.1038/s41366-021-01037-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Pannexin 3 (PANX3) is a channel-forming glycoprotein that enables nutrient-induced inflammation in vitro, and genetic linkage data suggest that it regulates body mass index. Here, we characterized inflammatory and metabolic parameters in global Panx3 knockout (KO) mice in the context of forced treadmill running (FEX) and high-fat diet (HFD). METHODS C57BL/6N (WT) and KO mice were randomized to either a FEX running protocol or no running (SED) from 24 until 30 weeks of age. Body weight was measured biweekly, and body composition was measured at 24 and 30 weeks of age. Male WT and KO mice were fed a HFD from 12 to 28 weeks of age. Metabolic organs were analyzed for a panel of inflammatory markers and PANX3 expression. RESULTS In females there were no significant differences in body composition between genotypes, which could be due to the lack of PANX3 expression in female white adipose tissue, while male KOs fed a chow diet had lower body weight and lower fat mass at 24 and 30 weeks of age, which was reduced to the same extent as 6 weeks of FEX in WT mice. In addition, male KO mice exhibited significantly lower expression of multiple pro-inflammatory genes in white adipose tissue compared to WT mice. While on a HFD body weight differences were insignificant, multiple inflammatory genes were significantly different in quadriceps muscle and white adipose tissue resulting in a more anti-inflammatory phenotype in KO mice compared to WT. The lower fat mass in male KO mice may be due to significantly fewer adipocytes in their subcutaneous fat compared to WT mice. Mechanistically, adipose stromal cells (ASCs) cultured from KO mice grow significantly slower than WT ASCs. CONCLUSION PANX3 is expressed in male adult mouse adipose tissue and may regulate adipocyte numbers, influencing fat accumulation and inflammation.
Collapse
Affiliation(s)
- C Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, ON, N6G 2V4, Canada
| | - Vanessa R Lee
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Danielle Johnston
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Nicolas J Pillon
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Samar Sayedyahossein
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Justin Tang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Rafael E Sanchez-Pupo
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Kevin J Barr
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Robert Gros
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, N6A 5C1, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Lauren Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, ON, N6G 2V4, Canada
- Department of Chemical and Biomedical Engineering, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Nica M Borradaile
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Frank Beier
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, ON, N6G 2V4, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, ON, N6G 2V4, Canada.
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
30
|
Aikawa Y, Yamashita T, Nakai N, Higashida K. Low-carbohydrate, high-fat diet and running exercise influence bone parameters in old mice. J Appl Physiol (1985) 2022; 132:1204-1212. [PMID: 35358401 DOI: 10.1152/japplphysiol.00789.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We examined the effects and interactions of a low-carbohydrate, high-fat (LCHF) diet and voluntary running exercise on bone in older mice. Male 19-month-old mice were divided into four groups by diet (control vs. LCHF) and exercise (sedentary vs. voluntary running). The control diet was 55% carbohydrate, 23% protein, and 22% fat, and the LCHF diet was 10% carbohydrate, 33% protein, and 57% fat as percentages of calories. The experiment ended when the mice reached 24 months old. Statistical analysis was conducted using two-way analysis of variance with diet and exercise. The LCHF diet decreased bone mineral content (BMC), bone mineral density, bone volume fraction, and trabecular number. There was no significant interaction between diet and exercise on many bone parameters. However, there were significant diet and exercise interactions on lumbar BMC and tibial trabecular total tissue volume and average cortical thickness. The LCHF diet attenuated the benefit of running exercise on lumbar BMC and caused running to have a negative effect on tibial trabecular total tissue volume. Our study suggests that the LCHF diet impairs bone mass and some trabecular microstructure and reduces the benefit of exercise on lumbar BMC in old mice.
Collapse
Affiliation(s)
- Yuki Aikawa
- Department of Food and Nutrition, Tsu City College, Tsu, Mie, Japan
| | - Takenori Yamashita
- Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Naoya Nakai
- Department of Nutrition, University of Shiga Prefecture, Hikone, Shiga, Japan
| | - Kazuhiko Higashida
- Department of Nutrition, University of Shiga Prefecture, Hikone, Shiga, Japan
| |
Collapse
|
31
|
The Nutrition-Microbiota-Physical Activity Triad: An Inspiring New Concept for Health and Sports Performance. Nutrients 2022; 14:nu14050924. [PMID: 35267899 PMCID: PMC8912693 DOI: 10.3390/nu14050924] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
The human gut microbiota is currently the focus of converging interest in many diseases and sports performance. This review presents gut microbiota as a real “orchestra conductor” in the host’s physio(patho)logy due to its implications in many aspects of health and disease. Reciprocally, gut microbiota composition and activity are influenced by many different factors, such as diet and physical activity. Literature data have shown that macro- and micro-nutrients influence gut microbiota composition. Cumulative data indicate that gut bacteria are sensitive to modulation by physical activity, as shown by studies using training and hypoactivity models. Sports performance studies have also presented interesting and promising results. Therefore, gut microbiota could be considered a “pivotal” organ for health and sports performance, leading to a new concept: the nutrition-microbiota-physical activity triad. The next challenge for the scientific and medical communities is to test this concept in clinical studies. The long-term aim is to find the best combination of the three elements of this triad to optimize treatments, delay disease onset, or enhance sports performance. The many possibilities offered by biotic supplementation and training modalities open different avenues for future research.
Collapse
|
32
|
Faraj M, Napoli N. The Impact of Diet on Bone and Fracture Risk in Diabetes. Curr Osteoporos Rep 2022; 20:26-42. [PMID: 35201556 DOI: 10.1007/s11914-022-00725-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the recently published scientific evidence on the effects of diet on diabetes and skeletal health. RECENT FINDINGS The impact of diet on overall health has been a growing topic of interest among researchers. An inappropriate eating habit is a relatively modified risk factor for diabetes in adults. Parallel with the significant increase in the incidence of diabetes mellitus worldwide, many studies have shown the benefits of lifestyle modifications, including diet and exercise for people with, or at risk of developing, diabetes. In the last years, accumulating evidence suggests that diabetes is a risk factor for bone fragility. As lifestyle intervention represents an effective option for diabetes management and treatment, there is potential for an effect on bone health. Healthy lifestyle is critical to prevent bone fragility. However, more studies are needed to fully understand the impact of diet and weight loss on fracture risk in diabetics.
Collapse
Affiliation(s)
- M Faraj
- Unit of Endocrinology and Diabetes, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128, Rome, Italy
| | - N Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128, Rome, Italy.
- Division of Bone and Mineral Diseases, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
33
|
Stauch CM, Fanburg-Smith JC, Walley KC, King JL, Murie B, Kim M, Koroneos Z, Waning D, Elfar JC, Aynardi MC. Animal model detects early pathologic changes of Charcot neuropathic arthropathy. Ann Diagn Pathol 2022; 56:151878. [PMID: 34953234 DOI: 10.1016/j.anndiagpath.2021.151878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 11/01/2022]
Abstract
Charcot neuropathic arthropathy is a degenerative, debilitating disease that affects the foot and ankle in patients with diabetes and peripheral neuropathy, often resulting in destruction, amputation. Proposed etiologies include neurotraumatic, inflammatory, and neurovascular. There has been no previous animal model for Charcot. This study proposes a novel rodent model of induced neuropathic arthropathy to understand the earliest progressive pathologic changes of human Charcot. High-fat-diet-induced obese (DIO) Wild-type C57BL/6J mice (n = 8, diabetic) and age-matched low-fat-diet controls (n = 6) were run on an inclined high-intensity treadmill protocol four times per week for 7 weeks to induce mechanical neurotrauma to the hind-paw, creating Charcot neuropathic arthropathy. Sensory function and radiologic correlation were assessed; animals were sacrificed to evaluate hindpaw soft tissue and joint pathology. With this model, Charcot-DIO mice reveals early pathologic features of Charcot neuropathic arthropathy, a distinctive subchondral microfracture callus, perichondral/subchondral osseous hypertrophy/osteosclerosis, that precedes fragmentation/destruction observed in human surgical pathology specimens. There is intraneural vacuolar-myxoid change and arteriolosclerosis. The DIO mice demonstrated significant hot plate sensory neuropathy compared (P < 0.01), radiographic collapse of the longitudinal arch in DIO mice (P < 0.001), and diminished bone density in DIO, compared with normal controls. Despite exercise, high-fat-DIO mice increased body weight and percentage of body fat (P < 0.001). This murine model of diet-induced obesity and peripheral neuropathy, combined with repetitive mechanical trauma, simulates the earliest changes observed in human Charcot neuropathic arthropathy, of vasculopathic-neuropathic etiology. An understanding of early pathophysiology may assist early diagnosis and intervention and reduce patient morbidity and mortality in Charcot neuropathic arthropathy.
Collapse
Affiliation(s)
- Christopher M Stauch
- Penn State College of Medicine, Milton S. Hershey Medical Center in Hershey, PA, United States of America; Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - Julie C Fanburg-Smith
- Penn State College of Medicine, Milton S. Hershey Medical Center in Hershey, PA, United States of America; Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America; Department of Pathology, Milton S. Hershey Medical Center in Hershey, PA, United States of America; Department of Pediatrics, Milton S. Hershey Medical Center in Hershey, PA, United States of America.
| | - Kempland C Walley
- Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - Jesse L King
- Penn State College of Medicine, Milton S. Hershey Medical Center in Hershey, PA, United States of America; Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - Benjamin Murie
- Department of Pathology, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - Morgan Kim
- Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - Zachary Koroneos
- Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - David Waning
- Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - John C Elfar
- Penn State College of Medicine, Milton S. Hershey Medical Center in Hershey, PA, United States of America; Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| | - Michael C Aynardi
- Penn State College of Medicine, Milton S. Hershey Medical Center in Hershey, PA, United States of America; Department of Orthopedics, Milton S. Hershey Medical Center in Hershey, PA, United States of America
| |
Collapse
|
34
|
Lu L, Tang M, Li J, Xie Y, Li Y, Xie J, Zhou L, Liu Y, Yu X. Gut Microbiota and Serum Metabolic Signatures of High-Fat-Induced Bone Loss in Mice. Front Cell Infect Microbiol 2022; 11:788576. [PMID: 35004355 PMCID: PMC8727351 DOI: 10.3389/fcimb.2021.788576] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/06/2021] [Indexed: 02/05/2023] Open
Abstract
Background Accumulating evidence indicates that high-fat diet (HFD) is a controllable risk factor for osteoporosis, but the underlying mechanism remains to be elucidated. As a primary biological barrier for nutrient entry into the human body, the composition and function of gut microbiota (GM) can be altered rapidly by HFD, which may trigger abnormal bone metabolism. In the current study, we analyzed the signatures of GM and serum metabolomics in HFD-induced bone loss and explored the potential correlations of GM and serum metabolites on HFD-related bone loss. Methods We conducted a mouse model with HFD-induced bone loss through a 12-week diet intervention. Micro-CT, Osmium-μCT, and histological analyses were used to observe bone microstructure and bone marrow adipose tissue. Quantitative Real-Time PCR was applied to analyze gene expression related to osteogenesis, adipogenesis, and osteoclastogenesis. Enzyme-linked immunosorbent assay was used to measure the biochemical markers of bone turnover. 16s rDNA sequencing was employed to analyze the abundance of GM, and UHPLC-MS/MS was used to identify serum metabolites. Correlation analysis was performed to explore the relationships among bone phenotypes, GM, and the metabolome. Results HFD induced bone loss accompanied by bone marrow adipose tissue expansion and bone formation inhibition. In the HFD group, the relative abundance of Firmicutes was increased significantly, while Bacteroidetes, Actinobacteria, Epsilonbacteraeota, and Patescibacteria were decreased compared with the ND group. Association analysis showed that thirty-two bacterial genera were significantly related to bone volume per tissue volume (BV/TV). One hundred and forty-five serum metabolites were identified as differential metabolites associated with HFD intervention, which were significantly enriched in five pathways, such as purine metabolism, regulation of lipolysis in adipocyte and cGMP-PKG signaling pathway. Sixty-four diffiential metabolites were matched to the MS2 spectra; and ten of them were positively correlated with BV/TV and five were negatively correlated with BV/TV. Conclusions These findings indicated that the alternations of GM and serum metabolites were related to HFD-induced bone loss, which might provide new insights into explain the occurrence and development of HFD-related osteoporosis. The regulatory effects of GM and metabolites associated with HFD on bone homeostasis required further exploration.
Collapse
Affiliation(s)
- Lingyun Lu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Department of Integrated Traditional Chinese and Western Medicine, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Mengjia Tang
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Li
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Xie
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yujue Li
- Department of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Jinwei Xie
- Department of Orthopaedic Surgery and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhou
- Core Facilities of West China Hospital, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Abolghasemi A, Manca C, Iannotti FA, Shen M, Leblanc N, Lacroix S, Martin C, Flamand N, Di Marzo V, Silvestri C. Assessment of the Effects of Dietary Vitamin D Levels on Olanzapine-Induced Metabolic Side Effects: Focus on the Endocannabinoidome-Gut Microbiome Axis. Int J Mol Sci 2021; 22:12361. [PMID: 34830242 PMCID: PMC8620071 DOI: 10.3390/ijms222212361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022] Open
Abstract
Vitamin D deficiency is associated with poor mental health and dysmetabolism. Several metabolic abnormalities are associated with psychotic diseases, which can be compounded by atypical antipsychotics that induce weight gain and insulin resistance. These side-effects may be affected by vitamin D levels. The gut microbiota and endocannabinoidome (eCBome) are significant regulators of both metabolism and mental health, but their role in the development of atypical antipsychotic drug metabolic side-effects and their interaction with vitamin D status is unknown. We studied the effects of different combinations of vitamin D levels and atypical antipsychotic drug (olanzapine) exposure on whole-body metabolism and the eCBome-gut microbiota axis in female C57BL/6J mice under a high fat/high sucrose (HFHS) diet in an attempt to identify a link between the latter and the different metabolic outputs induced by the treatments. Olanzapine exerted a protective effect against diet-induced obesity and insulin resistance, largely independent of dietary vitamin D status. These changes were concomitant with olanzapine-mediated decreases in Trpv1 expression and increases in the levels of its agonists, including various N-acylethanolamines and 2-monoacylglycerols, which are consistent with the observed improvement in adiposity and metabolic status. Furthermore, while global gut bacteria community architecture was not altered by olanzapine, we identified changes in the relative abundances of various commensal bacterial families. Taken together, changes of eCBome and gut microbiota families under our experimental conditions might contribute to olanzapine and vitamin D-mediated inhibition of weight gain in mice on a HFHS diet.
Collapse
Affiliation(s)
- Armita Abolghasemi
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Joint International Unit between the National Research Council (CNR) of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Université Laval, Québec, QC G1V 0A6, Canada
| | - Claudia Manca
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Joint International Unit between the National Research Council (CNR) of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Université Laval, Québec, QC G1V 0A6, Canada
| | - Fabio A. Iannotti
- Joint International Unit between the National Research Council (CNR) of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Institute of Biomolecular Chemistry, National Council of Research (Consiglio Nazionale delle Ricerche), 80087 Pozzuoli, Italy;
| | - Melissa Shen
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Nadine Leblanc
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada
| | - Sébastien Lacroix
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada
| | - Cyril Martin
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
| | - Nicolas Flamand
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Vincenzo Di Marzo
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Joint International Unit between the National Research Council (CNR) of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Université Laval, Québec, QC G1V 0A6, Canada
- Joint International Unit between the National Research Council (CNR) of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Institute of Biomolecular Chemistry, National Council of Research (Consiglio Nazionale delle Ricerche), 80087 Pozzuoli, Italy;
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada
- École de Nutrition, Faculté des Sciences de L’agriculture et de L’alimentation (FSAA), Université Laval, Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), Université Laval, Québec, QC G1V 0A6, Canada
| | - Cristoforo Silvestri
- Centre de Recherche, l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (A.A.); (C.M.); (M.S.); (N.L.); (S.L.); (C.M.); (N.F.); (V.D.M.)
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Joint International Unit between the National Research Council (CNR) of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Université Laval, Québec, QC G1V 0A6, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
36
|
Harahap IA, Suliburska J. Probiotics and Isoflavones as a Promising Therapeutic for Calcium Status and Bone Health: A Narrative Review. Foods 2021; 10:2685. [PMID: 34828966 PMCID: PMC8621960 DOI: 10.3390/foods10112685] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/21/2022] Open
Abstract
Probiotics have potential clinical effects for treating and preventing osteoporosis. Meanwhile, isoflavones have attracted much attention due to their ability to prevent postmenopausal symptoms. Research has established that probiotics and isoflavones can regulate hormones, immune cells, and the gastrointestinal system, acting as links in the gut-bone axis. However, combining the effects of probiotics and isoflavones on calcium status and bone health is a more novel and a still-evolving research area. Lactobacillus and Bifidobacterium are the foremost strains that influence bone health to a significant extent. Among the isoflavones, daidzein, genistein, and the metabolites of genistein (such as equol) stimulate bone formation. It can be concluded that probiotics and isoflavones promote bone health by regulating calcium uptake, gut microbiota, and various metabolic pathways that are associated with osteoblast activity and bone formation. Nevertheless, further experiments of probiotics and isoflavones are still necessary to confirm the association between calcium bioavailability and bone health.
Collapse
|
37
|
Huang T, Fu X, Wang N, Yang M, Zhang M, Wang B, Chen T, Majaz S, Wang H, Wong CW, Liu J, Guan M. Andrographolide prevents bone loss via targeting estrogen-related receptor-α-regulated metabolic adaption of osteoclastogenesis. Br J Pharmacol 2021; 178:4352-4367. [PMID: 34233019 DOI: 10.1111/bph.15614] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/27/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Metabolic adaptation driven by oestrogen-related receptor-α (ERRα/NR3B1) is required to meet the increased energy demand during osteoclast differentiation. Here, we hypothesize that natural product, andrographolide, acts as an ERRα inverse agonist to inhibit osteoclastogenesis. EXPERIMENTAL APPROACH Virtual docking and site-directed mutagenesis analysis were employed to study the binding mode of andrographolide to ERRα. Co-immunoprecipitation, luciferase reporter assay, real-time polymerase chain reaction (PCR) and immunoblot analyses were performed to identify andrographolide as an ERRα inverse agonist. The pharmacological effects of andrographolide in vivo were assessed in mice models of osteopenia induced by either a high-fat diet in male or ovariectomy in female mice. KEY RESULTS ERRα-dependent expression of glutaminase, a rate-limiting enzyme of mitochondrial glutamine anaplerosis, is required for ex vivo bone marrow osteoclast differentiation. Andrographolide inhibited glutaminase expression induced by ERRα and co-activator peroxisome proliferator-activated receptor γ co-activator-1β (PGC-1β), leading to reduction in osteoclastogenesis. Andrographolide acted as an inverse agonist of ERRα by disrupting its interaction with co-activator PGC-1β. Phenylalanine 232, valine 395 and phenylalanine 399 of ERRα ligand-binding domain were confirmed to be essential for this effect. In contrast, glutaminase overexpression restored the impairment triggered by andrographolide. Accordingly, andrographolide suppressed osteoclastic bone resorption and attenuated bone loss in vivo. CONCLUSIONS AND IMPLICATIONS These findings demonstrate that andrographolide acts as an ERRα inverse agonist for perturbation of ERRα/PGC-1β/glutaminase axis-driven metabolic adaption during osteoclast differentiation, implying that andrographolide may be a promising natural compound for preventing physiological and pathological bone loss.
Collapse
Affiliation(s)
- Tongling Huang
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuekun Fu
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Na Wang
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Meng Yang
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Minyi Zhang
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Binxu Wang
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Tianke Chen
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sidra Majaz
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chi-Wai Wong
- NeuMed Pharmaceuticals Limited, Yuen Long, Hong Kong, China
| | - Jinsong Liu
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Min Guan
- Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
38
|
Ravichandran A, Meinert C, Bas O, Hutmacher DW, Bock N. Engineering a 3D bone marrow adipose composite tissue loading model suitable for studying mechanobiological questions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112313. [PMID: 34474864 DOI: 10.1016/j.msec.2021.112313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Tissue engineering strategies are widely used to model and study the bone marrow microenvironment in healthy and pathological conditions. Yet, while bone function highly depends on mechanical stimulation, the effects of biomechanical stimuli on the bone marrow niche, specifically on bone marrow adipose tissue (BMAT) is poorly understood due to a lack of representative in vitro loading models. Here, we engineered a BMAT analog made of a GelMA (gelatin methacryloyl) hydrogel/medical-grade polycaprolactone (mPCL) scaffold composite to structurally and biologically mimic key aspects of the bone marrow microenvironment, and exploited an innovative bioreactor to study the effects of mechanical loading. Highly reproducible BMAT analogs facilitated the successful adipogenesis of human mesenchymal bone marrow stem cells. Upon long-term intermittent stimulation (1 Hz, 2 h/day, 3 days/week, 3 weeks) in the novel bioreactor, cellular proliferation and lipid accumulation were similar to unloaded controls, yet there was a significant reduction in the secretion of adipokines including leptin and adiponectin, in line with clinical evidence of reduced adipokine expression following exercise/activity. Ultimately, this innovative loading platform combined with reproducibly engineered BMAT analogs provide opportunities to study marrow physiology in greater complexity as it accounts for the dynamic mechanical microenvironment context.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia
| | - Christoph Meinert
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Metro North Hospital and Health Service, Herston 4029, QLD, Australia
| | - Onur Bas
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove 4059, QLD, Australia
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; Bone and Joint Disorders Program, School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane 4000, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia; ARC Industrial Transformation Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing, QUT, Kelvin Grove 4059, QLD, Australia.
| |
Collapse
|
39
|
Seely KD, Kotelko CA, Douglas H, Bealer B, Brooks AE. The Human Gut Microbiota: A Key Mediator of Osteoporosis and Osteogenesis. Int J Mol Sci 2021; 22:9452. [PMID: 34502371 PMCID: PMC8431678 DOI: 10.3390/ijms22179452] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/14/2022] Open
Abstract
An expanding body of research asserts that the gut microbiota has a role in bone metabolism and the pathogenesis of osteoporosis. This review considers the human gut microbiota composition and its role in osteoclastogenesis and the bone healing process, specifically in the case of osteoporosis. Although the natural physiologic processes of bone healing and the pathogenesis of osteoporosis and bone disease are now relatively well known, recent literature suggests that a healthy microbiome is tied to bone homeostasis. Nevertheless, the mechanism underlying this connection is still somewhat enigmatic. Based on the literature, a relationship between the microbiome, osteoblasts, osteoclasts, and receptor activator of nuclear factor-kappa-Β ligand (RANKL) is contemplated and explored in this review. Studies have proposed various mechanisms of gut microbiome interaction with osteoclastogenesis and bone health, including micro-RNA, insulin-like growth factor 1, and immune system mediation. However, alterations to the gut microbiome secondary to pharmaceutical and surgical interventions cannot be discounted and are discussed in the context of clinical therapeutic consideration. The literature on probiotics and their mechanisms of action is examined in the context of bone healing. The known and hypothesized interactions of common osteoporosis drugs and the human gut microbiome are examined. Since dysbiosis in the gut microbiota can function as a biomarker of bone metabolic activity, it may also be a pharmacological and nutraceutical (i.e., pre- and probiotics) therapeutic target to promote bone homeostasis.
Collapse
Affiliation(s)
- Kevin D. Seely
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Cody A. Kotelko
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Hannah Douglas
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Brandon Bealer
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
| | - Amanda E. Brooks
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA; (C.A.K.); (H.D.); (B.B.); (A.E.B.)
- Department of Research and Scholarly Activity, Rocky Vista University, Ivins, UT 84738, USA
| |
Collapse
|
40
|
MicroRNA-29a in Osteoblasts Represses High-Fat Diet-Mediated Osteoporosis and Body Adiposis through Targeting Leptin. Int J Mol Sci 2021; 22:ijms22179135. [PMID: 34502056 PMCID: PMC8430888 DOI: 10.3390/ijms22179135] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/14/2022] Open
Abstract
Skeletal tissue involves systemic adipose tissue metabolism and energy expenditure. MicroRNA signaling controls high-fat diet (HFD)-induced bone and fat homeostasis dysregulation remains uncertain. This study revealed that transgenic overexpression of miR-29a under control of osteocalcin promoter in osteoblasts (miR-29aTg) attenuated HFD-mediated body overweight, hyperglycemia, and hypercholesterolemia. HFD-fed miR-29aTg mice showed less bone mass loss, fatty marrow, and visceral fat mass together with increased subscapular brown fat mass than HFD-fed wild-type mice. HFD-induced O2 underconsumption, respiratory quotient repression, and heat underproduction were attenuated in miR-29aTg mice. In vitro, miR-29a overexpression repressed transcriptomic landscapes of the adipocytokine signaling pathway, fatty acid metabolism, and lipid transport, etc., of bone marrow mesenchymal progenitor cells. Forced miR-29a expression promoted osteogenic differentiation but inhibited adipocyte formation. miR-29a signaling promoted brown/beige adipocyte markers Ucp-1, Pgc-1α, P2rx5, and Pat2 expression and inhibited white adipocyte markers Tcf21 and Hoxc9 expression. The microRNA also reduced peroxisome formation and leptin expression during adipocyte formation and downregulated HFD-induced leptin expression in bone tissue. Taken together, miR-29a controlled leptin signaling and brown/beige adipocyte formation of osteogenic progenitor cells to preserve bone anabolism, which reversed HFD-induced energy underutilization and visceral fat overproduction. This study sheds light on a new molecular mechanism by which bone integrity counteracts HFD-induced whole-body fat overproduction.
Collapse
|
41
|
Tencerova M, Ferencakova M, Kassem M. Bone marrow adipose tissue: Role in bone remodeling and energy metabolism. Best Pract Res Clin Endocrinol Metab 2021; 35:101545. [PMID: 33966979 DOI: 10.1016/j.beem.2021.101545] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bone marrow adipose tissue (BMAT) has been considered for several decades as a silent bystander that fills empty space left in bone marrow following age-related decrease in hematopoiesis. However, recently new discoveries revealed BMAT as a secretory and metabolically active organ contributing to bone and whole-body energy metabolism. BMAT exhibits metabolic functions distinct from extramedullary adipose depots, relevant to its role in regulation of energy metabolism and its contribution to fracture risk observed in metabolic bone diseases. This review discusses novel insights of BMAT with particular emphasis on its contribution to the regulation of bone homeostasis. We also discuss the role of BMAT in regulation of fuel utilization and energy use that affect skeletal stem cell functions.
Collapse
Affiliation(s)
- Michaela Tencerova
- Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Michaela Ferencakova
- Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Moustapha Kassem
- Molecular Endocrinology and Stem Cell Research Unit, Department of Endocrinology and Metabolism, Odense University Hospital and Institute of Clinical Research, University of Southern Denmark, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Clauss M, Gérard P, Mosca A, Leclerc M. Interplay Between Exercise and Gut Microbiome in the Context of Human Health and Performance. Front Nutr 2021; 8:637010. [PMID: 34179053 PMCID: PMC8222532 DOI: 10.3389/fnut.2021.637010] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Gut microbiota and exercise have recently been shown to be interconnected. Both moderate and intense exercise are typically part of the training regimen of endurance athletes, but they exert different effects on health. Moderate exercise has positive effects on the health of average athletes, such as a reduction in inflammation and intestinal permeability and an improvement in body composition. It also induces positive changes in the gut microbiota composition and in the microbial metabolites produced in the gastrointestinal tract. Conversely, intense exercise can increase gastrointestinal epithelial wall permeability and diminish gut mucus thickness, potentially enabling pathogens to enter the bloodstream. This, in turn, may contribute to the increase in inflammation levels. However, elite athletes seem to have a higher gut microbial diversity, shifted toward bacterial species involved in amino acid biosynthesis and carbohydrate/fiber metabolism, consequently producing key metabolites such as short-chain fatty acids. Moreover, rodent studies have highlighted a bidirectional relationship, with exercise impacting the gut microbiota composition while the microbiota may influence performance. The present review focuses on gut microbiota and endurance sports and how this interconnection depends upon exercise intensity and training. After pointing out the limits of the studies so far available, we suggest that taking into account the microbiota composition and its metabolic contribution to human host health could help in monitoring and modulating athletes' health and performance. Such an integrated approach should help in the design of microbiome-based solutions for health or performance.
Collapse
Affiliation(s)
- Matthieu Clauss
- Université Paris-Saclay, INRAE, AgroParisTech, MICALIS Institute, Jouy-en-Josas, France
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Philippe Gérard
- Université Paris-Saclay, INRAE, AgroParisTech, MICALIS Institute, Jouy-en-Josas, France
| | - Alexis Mosca
- Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- Institut National de la Santé et de la Recherche Médicale et Université Paris Diderot, Sorbonne Paris-Cité, United Medical Resources 1149 Labex Inflamex, Paris, France
| | - Marion Leclerc
- Université Paris-Saclay, INRAE, AgroParisTech, MICALIS Institute, Jouy-en-Josas, France
| |
Collapse
|
43
|
Abstract
PURPOSE OF THE REVIEW The human gut harbors a complex community of microbes that influence many processes regulating musculoskeletal development and homeostasis. This review gives an update on the current knowledge surrounding the impact of the gut microbiota on musculoskeletal health, with an emphasis on research conducted over the last three years. RECENT FINDINGS The gut microbiota and their metabolites are associated with sarcopenia, osteoporosis, osteoarthritis, and rheumatoid arthritis. The field is moving fast from describing simple correlations to pursue establishing causation through clinical trials. The gut microbiota and their microbial-synthesized metabolites hold promise for offering new potential alternatives for the prevention and treatment of musculoskeletal diseases given its malleability and response to environmental stimuli.
Collapse
Affiliation(s)
- R Li
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - C G Boer
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - L Oei
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
44
|
Jensen VFH, Mølck AM, Dalgaard M, McGuigan FE, Akesson KE. Changes in bone mass associated with obesity and weight loss in humans: Applicability of animal models. Bone 2021; 145:115781. [PMID: 33285255 DOI: 10.1016/j.bone.2020.115781] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/05/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022]
Abstract
The implications of obesity and weight loss for human bone health are not well understood. Although the bone changes associated with weight loss are similar in humans and rodents, that is not the case for obesity. In humans, obesity is generally associated with increased bone mass, an outcome which is exacerbated by advanced age and menopause. In rodents, by contrast, bone mass decreases in proportion to severity and duration of obesity, and is influenced by sex, age and mechanical load. Despite these discrepancies, rodents are frequently used to model the situation in humans. In this review, we summarise the existing knowledge of the effects of obesity and weight loss on bone mass in humans and rodents, focusing on the translatability of findings from animal models. We then describe how animal models should be used to broaden the understanding of the relationship between obesity, weight loss, and skeletal health in humans. Specifically, we highlight the aspects of study design that should be considered to optimise translatability of the rodent models of obesity and weight loss. Notably, the sex, age, and nutritional status of the animals should ideally match those of interest in humans. With these caveats in mind, and depending on the research question asked, our review underscores that animal models can provide valuable information for obesity and weight-management research.
Collapse
Affiliation(s)
- Vivi F H Jensen
- Lund University, Department of Clinical Sciences Malmö and Skåne University Hospital, Department of Orthopedics, Inga Marie Nilssons Gata 22, 205 02 Malmö, Sweden.
| | - Anne-Marie Mølck
- Novo Nordisk A/S, Department of Safety Sciences, Imaging & Data Management, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Majken Dalgaard
- Novo Nordisk A/S, Department of Safety Sciences, Imaging & Data Management, Novo Nordisk Park 1, 2760 Maaloev, Denmark
| | - Fiona E McGuigan
- Lund University, Department of Clinical Sciences Malmö and Skåne University Hospital, Department of Orthopedics, Inga Marie Nilssons Gata 22, 205 02 Malmö, Sweden
| | - Kristina E Akesson
- Lund University, Department of Clinical Sciences Malmö and Skåne University Hospital, Department of Orthopedics, Inga Marie Nilssons Gata 22, 205 02 Malmö, Sweden
| |
Collapse
|
45
|
Aya V, Flórez A, Perez L, Ramírez JD. Association between physical activity and changes in intestinal microbiota composition: A systematic review. PLoS One 2021; 16:e0247039. [PMID: 33630874 PMCID: PMC7906424 DOI: 10.1371/journal.pone.0247039] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/31/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The intestinal microbiota comprises bacteria, fungi, archaea, protists, helminths and viruses that symbiotically inhabit the digestive system. To date, research has provided limited data on the possible association between an active lifestyle and a healthy composition of human microbiota. This review was aimed to summarize the results of human studies comparing the microbiome of healthy individuals with different physical activity amounts. METHODS We searched Medline/Ovid, NIH/PubMed, and Academic Search Complete between August-October 2020. Inclusion criteria comprised: (a) cross-sectional studies focused on comparing gut microbiome among subjects with different physical activity levels; (b) studies describing human gut microbiome responses to any type of exercise stimulus; (c) studies containing healthy adult women and men. We excluded studies containing diet modifications, probiotic or prebiotic consumption, as well as studies focused on diabetes, hypertension, cancer, hormonal dysfunction. Methodological quality and risk of bias for each study were assessed using the Risk Of Bias In Non-randomized Studies-of Interventions tool. The results from cross-sectional and longitudinal studies are shown independently. RESULTS A total of 17 articles were eligible for inclusion: ten cross-sectional and seven longitudinal studies. Main outcomes vary significantly according to physical activity amounts in longitudinal studies. We identified discrete changes in diversity indexes and relative abundance of certain bacteria in active people. CONCLUSION As literature in this field is rapidly growing, it is important that studies incorporate diverse methods to evaluate other aspects related to active lifestyles such as sleep and dietary patterns. Exploration of other groups such as viruses, archaea and parasites may lead to a better understanding of gut microbiota adaptation to physical activity and sports and its potentially beneficial effects on host metabolism and endurance.
Collapse
Affiliation(s)
- Viviana Aya
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Alberto Flórez
- Grupo In-Novum Educatio, Facultad de Educación, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Luis Perez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
46
|
Ni JJ, Yang XL, Zhang H, Xu Q, Wei XT, Feng GJ, Zhao M, Pei YF, Zhang L. Assessing causal relationship from gut microbiota to heel bone mineral density. Bone 2021; 143:115652. [PMID: 32971307 DOI: 10.1016/j.bone.2020.115652] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 12/28/2022]
Abstract
Recent studies have demonstrated the important role played by gut microbiota in regulating bone development, but the evidence of such causal relationship is still sparse in human population. The aim of this study is to assess the causal relationship from gut microbiota to bone development and to identify specific causal bacteria taxa via a Mendelian randomization (MR) approach. A genome-wide association study (GWAS) summary statistic based two-sample MR analysis was performed. Summary statistics of microbiome GWAS (MGWAS) in 1126 twin pairs of the TwinsUK study was used as discovery sample, and the MGWAS in 984 Dutch participants from the LifeLines-DEEP cohort was used as replication sample. Estimated heel bone mineral density (eBMD) GWAS in 426,824 participants from the UK biobank (UKB) cohort was used as outcome. Bacteria were grouped into taxa features at both order and family levels. In the discovery sample, a total of 25 bacteria features including 9 orders and 16 families were analyzed. Fourteen features (5 orders + 9 families) were nominally significant, including 5 orders (Bacteroidales, Clostridiales, Lactobacillales, Pasteurellales and Verrucomicrobiales) and 9 families (Bacteroidaceae, Clostridiaceae, Lachnospiraceae, Mogibacteriaceae, Pasteurellaceae, Porphyromonadaceae, Streptococcaceae, Verrucomicrobiaceae and Veillonellaceae). One order Clostridiales and its child taxon, family Lachnospiraceae, were successfully replicated in the replication sample (Clostridiales Pdiscovery = 3.32 × 10-3Preplication = 7.29 × 10-3; Lachnospiraceae Pdiscovery = 0.03 Preplication = 7.29 × 10-3). Our findings provided evidence of causal relationship from microbiota to bone development, as well as identified specific bacteria taxa that regulated bone mass variation, thus providing new insights into the microbiota mediated bone development mechanism.
Collapse
Affiliation(s)
- Jing-Jing Ni
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Xiao-Lin Yang
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Jiangsu, PR China; Laboratory of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Jiangsu, PR China
| | - Hong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Qian Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Xin-Tong Wei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Gui-Juan Feng
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Min Zhao
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Yu-Fang Pei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China.
| | - Lei Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China.
| |
Collapse
|
47
|
Hair R, Sakaki JR, Chun OK. Anthocyanins, Microbiome and Health Benefits in Aging. Molecules 2021; 26:537. [PMID: 33494165 PMCID: PMC7864342 DOI: 10.3390/molecules26030537] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/09/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
The percentage of individuals over the age of 60 is projected to reach 22% by 2050; chronic diseases associated with aging can present challenges for these individuals. Anthocyanins and the gut microbiome have each been studied as independent influencers of health. Both these factors have shown to have a positive effect on cardiovascular and bone health in individuals, as well as on the prevention or treatment of certain forms of cancers. Anthocyanins have shown to modulate the composition of the gut microbiome and may have overlapping mechanisms in the prevention and treatment of cardiovascular disease, cancer, neurodegenerative disorders and aging-associated bone loss. These health outcomes are responsible for the hospitalization and deaths of millions of Americans every year and they cost the United States billions of dollars each year to maintain, prevent and treat. Alternative methods of treatment and prevention are desired since conventional methods (surgical and pharmacological methods, physical therapy, etc.) can be costly and have significant side effects; evidence suggests that anthocyanins and the gut microbiome may be potential avenues for this. This review evaluates the findings of existing literature on the role of anthocyanins and the gut microbiome on health and their potential as a natural therapeutic agent or a target organ to provide an alternative to the conventional methods of disease prevention and treatment.
Collapse
Affiliation(s)
| | | | - Ock K. Chun
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA; (R.H.); (J.R.S.)
| |
Collapse
|
48
|
Qiao J, Wu Y, Ren Y. The impact of a high fat diet on bones: potential mechanisms. Food Funct 2021; 12:963-975. [PMID: 33443523 DOI: 10.1039/d0fo02664f] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High-fat diet led to bone loss via gut microbiota and fatty acid imbalances, immune disorder and adipose tissue accumulation inside and outside the bone marrow.
Collapse
Affiliation(s)
- Jie Qiao
- Department of Endocrinology and Metabolism
- the Second Affiliated Hospital of Zhejiang University School of Medicine
- Hangzhou
- 310009
- China
| | - Yiwen Wu
- Department of Neurosurgery
- Ningbo Hospital
- Zhejiang University School of Medicine
- Ningbo 315010
- China
| | - Yuezhong Ren
- Department of Endocrinology and Metabolism
- the Second Affiliated Hospital of Zhejiang University School of Medicine
- Hangzhou
- 310009
- China
| |
Collapse
|
49
|
Little-Letsinger SE, Pagnotti GM, McGrath C, Styner M. Exercise and Diet: Uncovering Prospective Mediators of Skeletal Fragility in Bone and Marrow Adipose Tissue. Curr Osteoporos Rep 2020; 18:774-789. [PMID: 33068251 PMCID: PMC7736569 DOI: 10.1007/s11914-020-00634-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW To highlight recent basic, translational, and clinical works demonstrating exercise and diet regulation of marrow adipose tissue (MAT) and bone and how this informs current understanding of the relationship between marrow adiposity and musculoskeletal health. RECENT FINDINGS Marrow adipocytes accumulate in the bone in the setting of not only hypercaloric intake (calorie excess; e.g., diet-induced obesity) but also with hypocaloric intake (calorie restriction; e.g., anorexia), despite the fact that these states affect bone differently. With hypercaloric intake, bone quantity is largely unaffected, whereas with hypocaloric intake, bone quantity and quality are greatly diminished. Voluntary running exercise in rodents was found to lower MAT and promote bone in eucaloric and hypercaloric states, while degrading bone in hypocaloric states, suggesting differential modulation of MAT and bone, dependent upon whole-body energy status. Energy status alters bone metabolism and bioenergetics via substrate availability or excess, which plays a key role in the response of bone and MAT to mechanical stimuli. Marrow adipose tissue (MAT) is a fat depot with a potential role in-as well as responsivity to-whole-body energy metabolism. Understanding the localized function of this depot in bone cell bioenergetics and substrate storage, principally in the exercised state, will aid to uncover putative therapeutic targets for skeletal fragility.
Collapse
Affiliation(s)
- Sarah E Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA.
| | - Gabriel M Pagnotti
- Department of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Cody McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
50
|
A Multi-Omic Analysis for Low Bone Mineral Density in Postmenopausal Women Suggests a RELATIONSHIP between Diet, Metabolites, and Microbiota. Microorganisms 2020; 8:microorganisms8111630. [PMID: 33105628 PMCID: PMC7690388 DOI: 10.3390/microorganisms8111630] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023] Open
Abstract
The effect of microbiota composition and its health on bone tissue is a novel field for research. However, their associations with bone mineral density (BMD) have not been established in postmenopausal women. The present study investigates the relation of diet, the microbiota composition, and the serum metabolic profile in postmenopausal women with normal-BMD or with low-BMD. Ninety-two Mexican postmenopausal women were classified into normal-BMD (n = 34) and low-BMD (n = 58). The V4 hypervariable region was sequenced using the Miseq platform. Serum vitamin D was determined by chemiluminescence immunoassay. Serum concentrations of acyl-carnitines and amino acids were determined by electrospray tandem mass spectrometry. Diet was assessed by a food frequency questionnaire. The low-BMD group had fewer observed species, higher abundance of γ-Proteobacteria, lower consumption of lycopene, and lower concentrations of leucine, valine, and tyrosine compared with the normal-BMD group. These amino acids correlated positively with the abundance of Bacteroides. Lycopene consumption positively correlated with Oscillospira and negatively correlated with Pantoea genus abundance. Finally, the intestinal microbiota of women with vitamin D deficiency was related to Erysipelotrichaceae and Veillonellaceae abundance compared to the vitamin D non-deficient group. Associations mediated by the gut microbiota between diet and circulating metabolites with low-BMD were identified.
Collapse
|