1
|
Liu Q, Xue Y, Guo J, Tao L, Zhu Y. Citrate: a key signalling molecule and therapeutic target for bone remodeling disorder. Front Endocrinol (Lausanne) 2025; 15:1512398. [PMID: 39886032 PMCID: PMC11779597 DOI: 10.3389/fendo.2024.1512398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/20/2024] [Indexed: 02/01/2025] Open
Abstract
Bone remodeling is a continuous cyclic process that maintains and regulates bone structure and strength. The disturbance of bone remodeling leads to a series of bone metabolic diseases. Recent studies have shown that citrate, an intermediate metabolite of the tricarboxylic acid (TCA) cycle, plays an important role in bone remodeling. But the exact mechanism is still unclear. In this study, we focused on the systemic regulatory mechanism of citrate on bone remodeling, and found that citrate is involved in bone remodeling in multiple ways. The participation of citrate in oxidative phosphorylation (OXPHOS) facilitates the generation of ATP, thereby providing substantial energy for bone formation and resorption. Osteoclast-mediated bone resorption releases citrate from bone mineral salts, which is subsequently released as an energy source to activate the osteogenic differentiation of stem cells. Finally, the differentiated osteoblasts secrete into the bone matrix and participate in bone mineral salts formation. As a substrate of histone acetylation, citrate regulates the expression of genes related to bone formation and bone reabsorption. Citrate is also a key intermediate in the metabolism and synthesis of glucose, fatty acids and amino acids, which are three major nutrients in the organism. Citrate can also be used as a biomarker to monitor bone mass transformation and plays an important role in the diagnosis and therapeutic evaluation of bone remodeling disorders. Citrate imbalance due to citrate transporter could result in the supression of osteoblast/OC function through histone acetylation, thereby contributing to disorders in bone remodeling. Therefore, designing drugs targeting citrate-related proteins to regulate bone citrate content provides a new direction for the drug treatment of diseases related to bone remodeling disorders.
Collapse
Affiliation(s)
| | | | | | - Lin Tao
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Yue Zhu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Liu Z, Mao Y, Yang K, Wang S, Zou F. A trend of osteocalcin in diabetes mellitus research: bibliometric and visualization analysis. Front Endocrinol (Lausanne) 2025; 15:1475214. [PMID: 39872315 PMCID: PMC11769813 DOI: 10.3389/fendo.2024.1475214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
Background Osteocalcin has attracted attention for its potential role in diabetes management. However, there has been no bibliometric assessment of scientific progress in this field. Methods We analysed 1680 articles retrieved from the Web of Science Core Collection (WoSCC) between 1 January 1986 and 10 May 2024 using various online tools. Result These papers accumulated 42,714 citations,with an average of 25.43 citations per paper. Publication output increased sharply from 1991 onwards. The United States and China are at the forefront of this research area. Discussion The keywords were grouped into four clusters: 'Differential and functional osteocalcin genes', 'Differential expression of osteocalcin genes in relation to diabetes mellitus', 'Role of osteocalcin in the assessment of osteoporosis and diabetes mellitus', and 'Indirect involvement of osteocalcin in metabolic processes'. Analysis using the VoS viewer suggests a shift in research focus towards the correlation between osteocalcin levels and diabetic complications, the clinical efficacy of therapeutic agents or vitamins in the treatment of osteoporosis in diabetic patients, and the mechanisms by which osteocalcin modulates insulin action. The proposed focus areas are "osteocalcin genes", "insulin regulation and osteoporosis ", "different populations", "diabetes-related complications" and "type 2 diabetes mellitus","effect of osteocalcin expression on insulin sensitivity as well as secretion","osteocalcin expression in different populations of diabetic patients and treatment-related studies".
Collapse
Affiliation(s)
- Zixu Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Yuchen Mao
- The First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Kangping Yang
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Shukai Wang
- The First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Fang Zou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Xiang T, Yang C, Deng Z, Sun D, Luo F, Chen Y. Krüppel-like factors family in health and disease. MedComm (Beijing) 2024; 5:e723. [PMID: 39263604 PMCID: PMC11387732 DOI: 10.1002/mco2.723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Krüppel-like factors (KLFs) are a family of basic transcription factors with three conserved Cys2/His2 zinc finger domains located in their C-terminal regions. It is acknowledged that KLFs exert complicated effects on cell proliferation, differentiation, survival, and responses to stimuli. Dysregulation of KLFs is associated with a range of diseases including cardiovascular disorders, metabolic diseases, autoimmune conditions, cancer, and neurodegenerative diseases. Their multidimensional roles in modulating critical pathways underscore the significance in both physiological and pathological contexts. Recent research also emphasizes their crucial involvement and complex interplay in the skeletal system. Despite the substantial progress in understanding KLFs and their roles in various cellular processes, several research gaps remain. Here, we elucidated the multifaceted capabilities of KLFs on body health and diseases via various compliable signaling pathways. The associations between KLFs and cellular energy metabolism and epigenetic modification during bone reconstruction have also been summarized. This review helps us better understand the coupling effects and their pivotal functions in multiple systems and detailed mechanisms of bone remodeling and develop potential therapeutic strategies for the clinical treatment of pathological diseases by targeting the KLF family.
Collapse
Affiliation(s)
- Tingwen Xiang
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Chuan Yang
- Department of Biomedical Materials Science Third Military Medical University (Army Medical University) Chongqing China
| | - Zihan Deng
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Dong Sun
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Fei Luo
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Yueqi Chen
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
- Department of Orthopedics Chinese PLA 76th Army Corps Hospital Xining China
| |
Collapse
|
4
|
Chuang TL, Koo M, Wang YF. The impact of diabetes, anemia, and renal function in the relationship between osteoporosis and fasting blood glucose among Taiwanese women: a cross-sectional study. BMC Womens Health 2024; 24:23. [PMID: 38172731 PMCID: PMC10765617 DOI: 10.1186/s12905-023-02851-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The aim of this study was to investigate the association between fasting blood glucose and osteoporosis in women with diabetes, anemia, and renal function. METHODS The medical records of women who underwent a general health examination at a regional hospital in southern Taiwan were retrospectively reviewed. Logistic regression analysis was performed to assess the association between osteoporosis and fasting blood glucose separately for the eight subgroups (diabetes or non-diabetes, anemia or non-anemia, normal or decreased renal function), adjusting for other clinical characteristics and laboratory findings. RESULTS A total of 11,872 women were included in the study. Among women with diabetes, anemia, and decreased renal function, an increment of 10 mg/dL in fasting blood glucose was associated with an increased risk of osteoporosis (adjusted odds ratio [aOR] = 1.57, p = 0.004). Among women without diabetes, fasting blood glucose was significantly associated with an increased risk of osteoporosis in those with anemia and normal renal function (OR = 1.14, p = 0.023) and those without anemia and normal renal function (OR = 1.04, p = 0.015), but these associations were not significant after adjusting for other covariates. CONCLUSIONS Higher fasting blood glucose levels in women with diabetes, anemia, and decreased renal function were associated with an increased risk of osteoporosis. Clinicians should be vigilant about glucose control in patients with diabetes to reduce the risk of fracture.
Collapse
Affiliation(s)
- Tzyy-Ling Chuang
- Department of Nuclear Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Malcolm Koo
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien, 970302, Taiwan.
| | - Yuh-Feng Wang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, No.201, Sec. 2, Shipai Road, Beitou District, Taipei City, 11217, Taiwan.
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Medical Imaging and Radiological Technology, Yuanpei University of Medical Technology, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Chen PW, Su HY, Tu YK, Wu CH, Yeh JI, Chen LY, Peng CCH, Loh CH, Huang HK, Lin SM. Association of bisphosphonates with diabetes risk and glycemic control: a meta-analysis. Osteoporos Int 2023; 34:387-397. [PMID: 36464699 DOI: 10.1007/s00198-022-06616-3] [Citation(s) in RCA: 4] [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: 07/22/2022] [Accepted: 11/16/2022] [Indexed: 12/07/2022]
Abstract
UNLABELLED Previous evidence suggests that bisphosphonates may improve glycemic control. The present meta-analysis, comprising seven studies with 1,233,844 participants, demonstrated that bisphosphonate use was significantly associated with a lower risk of diabetes. However, in the randomized controlled trial subgroup, a non-significant association was found. Further studies are needed to determine causality. PURPOSE This study aimed to evaluate the impact of bisphosphonates on glycemic control and the risk of incident diabetes. METHODS MEDLINE, Embase, and Cochrane Library were searched from inception to February 15, 2022. Experimental or observational studies that compared fasting blood glucose (FBG) and glycated hemoglobin (HbA1c) levels and the diabetes risk with and without bisphosphonates were included. Studies without relevant outcomes, only providing crude estimates, or the absence of a control group were excluded. Two reviewers independently screened the articles, extracted data, and appraised studies. The pooled relative risk (RR) and weighted mean difference (WMD) were calculated using random effects models. RESULTS Seven studies (n = 1,233,844) on diabetes risk were included, including two post hoc analyses of randomized controlled trials (RCTs) and five observational studies. Compared with controls, bisphosphonates (BPs) were associated with a significant decrease in the risk of diabetes (RR = 0.77; 95% CI, 0.65 to 0.90; P = 0.002). However, in the subgroup of post hoc analyses of RCTs, the association was non-significant (RR = 0.93; 95% CI, 0.74 to 1.18; P = 0.576). Moreover, three studies (n = 4906) on FBG and one (n = 60) on HbA1c were included. We observed non-significant association between BPs and changes in FBG (WMD = - 0.61 mg/dL; 95% CI, - 2.72 to 1.49; P = 0.567) and HbA1c (WMD = - 0.11%; 95% CI, - 0.23 to 0.01; P = 0.083). CONCLUSION Patients taking BPs may have a lower risk of incident diabetes than those without BPs. However, due to the high between-study heterogeneity and inconsistent findings between post hoc analyses of RCTs and observational studies, further rigorous RCTs are required to determine whether the findings are causal.
Collapse
Affiliation(s)
- Po-Wei Chen
- Department of Physical Medicine and Rehabilitation, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., 97002, Hualien, Taiwan
- Medical Department, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., 97002, Hualien, Taiwan
| | - Hsiao-Yuan Su
- Department of Psychiatric, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., Hualien, 97002, Taiwan
| | - Yu-Kang Tu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital and School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsing Wu
- Department of Family Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Family Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Gerontology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jih-I Yeh
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Family Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., Hualien, 97002, Taiwan
| | - Li-Yu Chen
- Library, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., Hualien, 97002, Taiwan
| | - Carol Chiung-Hui Peng
- Section of Endocrinology, Diabetes, Nutrition & Weight Management, Boston University School of Medicine, MA, Boston, USA
| | - Ching-Hui Loh
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Center for Aging and Health, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., Hualien, 97002, Taiwan
| | - Huei-Kai Huang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.
- School of Medicine, Tzu Chi University, Hualien, Taiwan.
- Department of Family Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., Hualien, 97002, Taiwan.
| | - Shu-Man Lin
- Department of Physical Medicine and Rehabilitation, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 707, Sec. 3, Chung Yang Rd., 97002, Hualien, Taiwan.
- School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
6
|
Chougule A, Baroi S, Czernik PJ, Crowe E, Chang MR, Griffin PR, Lecka-Czernik B. Osteocytes contribute via nuclear receptor PPAR-alpha to maintenance of bone and systemic energy metabolism. Front Endocrinol (Lausanne) 2023; 14:1145467. [PMID: 37181042 PMCID: PMC10173151 DOI: 10.3389/fendo.2023.1145467] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/20/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction The view that bone and energy metabolism are integrated by common regulatory mechanisms is broadly accepted and supported by multiple strands of evidence. This includes the well-characterized role of the PPARγ nuclear receptor, which is a common denominator in energy metabolism and bone metabolism. Little is known, however, about the role of PPARα nuclear receptor, a major regulator of lipid metabolism in other organs, in bone. Methods A side-by-side comparative study of 5-15 mo old mice with global PPARα deficiency (αKO) and mice with osteocyte-specific PPARα deficiency (αOTKO) in order to parse out the various activities of PPARα in the skeleton that are of local and systemic significance. This study included transcriptome analysis of PPARα-deficient osteocytes, and analyses of bone mass and bone microarchitecture, systemic energy metabolism with indirect calorimetry, and differentiation potential of hematopoietic and mesenchymal bone cell progenitors. These analyses were paired with in vitro studies of either intact or silenced for PPARα MLO-A5 cells to determine PPARα role in osteocyte bioenergetics. Results In osteocytes, PPARα controls large number of transcripts coding for signaling and secreted proteins which may regulate bone microenvironment and peripheral fat metabolism. In addition, PPARα in osteocytes controls their bioenergetics and mitochondrial response to stress, which constitutes up to 40% of total PPARα contribution to the global energy metabolism. Similarly to αKO mice, the metabolic phenotype of αOTKO mice (both males and females) is age-dependent. In younger mice, osteocyte metabolism contributes positively to global energetics, however, with aging the high-energy phenotype reverts to a low-energy phenotype and obesity develops, suggesting a longitudinal negative effect of impaired lipid metabolism and mitochondrial dysfunction in osteocytes deficient in PPARα. However, bone phenotype was not affected in αOTKO mice except in the form of an increased volume of marrow adipose tissue in males. In contrast, global PPARα deficiency in αKO mice led to enlarged bone diameter with a proportional increase in number of trabeculae and enlarged marrow cavities; it also altered differentiation of hematopoietic and mesenchymal marrow cells toward osteoclast, osteoblast and adipocyte lineages, respectively. Discussion PPARα role in bone is multileveled and complex. In osteocytes, PPARα controls the bioenergetics of these cells, which significantly contributes to systemic energy metabolism and their endocrine/paracrine function in controlling marrow adiposity and peripheral fat metabolism.
Collapse
Affiliation(s)
- Amit Chougule
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Sudipta Baroi
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Piotr J. Czernik
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Emily Crowe
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Mi Ra Chang
- Department of Molecular Medicine, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, United States
| | - Patrick R. Griffin
- Department of Molecular Medicine, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, United States
| | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- *Correspondence: Beata Lecka-Czernik,
| |
Collapse
|
7
|
Hu Z, Zhi X, Li J, Li B, Wang J, Zhu J, Zhang Z. Effects of long-term vitamin D supplementation on metabolic profile in middle-aged and elderly patients with type 2 diabetes. J Steroid Biochem Mol Biol 2023; 225:106198. [PMID: 36181990 DOI: 10.1016/j.jsbmb.2022.106198] [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: 06/28/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 02/01/2023]
Abstract
To evaluate the effects of long-term vitamin D supplementation on metabolic profiles in middle-aged to elderly patients with type 2 diabetes (T2D), a randomized controlled trial was conducted among patients with T2D aged 50-70 years. A total of 270 patients underwent randomization with 135 being allocated to the vitamin D group and 135 to the control group, and participants in the vitamin D group received oral vitamin D3 (800 IU/day) for 30 months. Serum 25(OH)D and metabolic variables were measured at baseline, and after 6, 12, 18, and 30 months of intervention. After 30 months, the vitamin D group showed a greater increase in serum 25(OH)D than the control group (12.39 ± 6.99 vs 5.35 ± 5.29 ng/ml, P < 0.001). Meanwhile, changes in the levels of fasting insulin, HOMA-IR, non-high-density-lipoprotein cholesterol (non-HDL-C), high-sensitivity C-reactive protein (hs-CRP), and uric acid differed significantly between the two groups (all P < 0.05). Stratified analysis indicated that change in uric acid differed significantly between the two groups in subgroup with baseline 25(OH)D ≥ 20 ng/ml (P = 0.042) or subgroup with female patients (P = 0.034). And the change in fasting blood glucose (FBG) differed significantly between the vitamin D group (-0.30 ± 2.52 mmol/L) and the control group (0.49 ± 1.78 mmol/L, P = 0.049) among patients achieving 25(OH)D concentrations of 30 ng/ml at the end of this trial. A significant difference in the change of triglyceride was observed between the two groups among patients with obesity at baseline [0.05(-0.59, 0.23) vs 0.41(-0.01, 0.80) mmol/L, P = 0.023]. These findings suggested that long-term vitamin D supplementation significantly reduced fasting insulin, HOMA-IR, and serum concentrations of non-HDL-C, hs-CRP, and uric acid among middle-aged to elderly patients with T2D. And vitamin D status, gender, and baseline obesity may modify the effects of vitamin D supplementation.
Collapse
Affiliation(s)
- Zhiyong Hu
- School of Public Health and Management, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, China; Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Xueyuan Zhi
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Jiafu Li
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Bingyan Li
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Jinxiu Wang
- School of Public Health and Management, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, China
| | - Jianliang Zhu
- Lishui Center for Disease Control and Prevention, Lishui 323000, China
| | - Zengli Zhang
- Department of Occupational and Environmental Health, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China.
| |
Collapse
|
8
|
Wang P, Zhang Y, Shan R, Wu J, Man S, Deng Y, Lv J, Wang X, Yin J, Ning Y, Wang B, Li L. Association between trajectories of fasting plasma glucose and risk of osteoporosis in non-diabetic and diabetic populations. Front Public Health 2022; 10:960928. [PMID: 36424968 PMCID: PMC9679646 DOI: 10.3389/fpubh.2022.960928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/12/2022] [Indexed: 11/11/2022] Open
Abstract
Introduction Previous studies based on a single measure of fasting plasma glucose (FPG) showed an inconsistent conclusion about the association between FPG and osteoporosis risk. Not accounting for time-varying and cumulative average of FPG over time could bias the true relation between FPG and osteoporosis. Our study aims to investigate the association between the trajectories of FPG and osteoporosis risk for non-diabetic and diabetic populations. Methods A total of 18,313 participants who attended physical examinations during 2008-2018 were included. They were free of osteoporosis at their first physical examination and followed until their last physical examination before December 31, 2018. We recorded their incidence of osteoporosis and at least three FPG values during follow-up. Their longitudinal FPG trajectories were identified by the latent class growth analysis model based on the changes in FPG. Multivariable logistic regression models were used to analyze the association between the trajectories of FPG and osteoporosis diagnosed in the follow-up physical examination in both non-diabetics and diabetics. Results There were 752 incident osteoporosis among 16,966 non-diabetic participants, and 57 incident osteoporosis among 1,347 diabetic participants. Among non-diabetics, the elevated-increasing FPG trajectory was negatively associated with osteoporosis risk in women (odds ratio (OR), 0.62; 95% confidence interval (CI), 0.43-0.88). Premenopausal women with elevated-increasing FPG trajectory had lower osteoporosis risk than those women with normal-stable FPG trajectory (OR, 0.41; 95% CI, 0.20-0.88), while this association was insignificant in postmenopausal women. Among diabetics, those whose longitudinal FPG is kept at a very high level had the highest risk of osteoporosis (OR, 3.09; 95% CI, 1.16-8.22), whereas those whose FPG starts with the high level and keeps on increasing did not exhibit a significantly increased risk (OR, 1.75; 95% CI, 0.81-3.76) compared with those who keep stable moderate-high level of FPG, except in men (OR, 2.49; 95% CI, 1.02-6.12). Conclusion Distinct trajectories of FPG are associated with differential risk of osteoporosis in non-diabetic and diabetic populations. Controlling a proper FPG level in different populations is necessary for osteoporosis prevention.
Collapse
Affiliation(s)
- Ping Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China,Department of Statistics and Information, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Yuanfeng Zhang
- Department of Medical Innovation, China Science and Technology Development Center for Chinese Medicine, Beijing, China
| | - Ruiqi Shan
- Department of Evidence Based Medicine, Meinian Institute of Health, Beijing, China,Peking University Health Science Center, Meinian Public Health Institute, Beijing, China
| | - Jing Wu
- Department of Evidence Based Medicine, Meinian Institute of Health, Beijing, China,Peking University Health Science Center, Meinian Public Health Institute, Beijing, China
| | - Sailimai Man
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China,Department of Evidence Based Medicine, Meinian Institute of Health, Beijing, China,Peking University Health Science Center, Meinian Public Health Institute, Beijing, China
| | - Yuhan Deng
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China,Peking University Health Science Center, Meinian Public Health Institute, Beijing, China,Department of Noncommunicable Diseases, Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Xiaona Wang
- Beijing MJ Health Screening Center Co., Ltd., Beijing, China
| | - Jianchun Yin
- Beijing MJ Health Screening Center Co., Ltd., Beijing, China
| | - Yi Ning
- Department of Evidence Based Medicine, Meinian Institute of Health, Beijing, China,School of Public Health, Hainan Medical University, Hainan, China,Yi Ning
| | - Bo Wang
- Department of Evidence Based Medicine, Meinian Institute of Health, Beijing, China,Peking University Health Science Center, Meinian Public Health Institute, Beijing, China,Bo Wang
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China,Peking University Health Science Center, Meinian Public Health Institute, Beijing, China,Department of Noncommunicable Diseases, Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China,*Correspondence: Liming Li
| |
Collapse
|
9
|
Lei H, Liu J, Wang W, Yang X, Feng Z, Zang P, Lu B, Shao J. Association between osteocalcin, a pivotal marker of bone metabolism, and secretory function of islet beta cells and alpha cells in Chinese patients with type 2 diabetes mellitus: an observational study. Diabetol Metab Syndr 2022; 14:160. [PMID: 36307866 PMCID: PMC9615358 DOI: 10.1186/s13098-022-00932-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/13/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Several recent studies have found that Osteocalcin (OCN), a multifunctional protein secreted exclusively by osteoblasts, is beneficial to glucose metabolism and type 2 diabetes mellitus (T2DM). However, the effects of OCN on islets function especially islet ɑ cells function in patients with type 2 diabetes mellitus characterized by a bi-hormonal disease are still unclear. The purpose of this cross-sectional study was to investigate the relationship between serum OCN and the secretion of islet β cells and ɑ cells in Chinese patients with type 2 diabetes mellitus. METHODS 204 patients with T2DM were enrolled. Blood glucose (FBG, PBG0.5h, PBG1h, PBG2h, PBG3h), insulin (FINS, INS0.5h, INS1h, INS2h, INS3h), C-peptide (FCP, CP0.5h, CP1h, CP2h, CP3h), and glucagon (GLA0, GLA0.5 h, GLA1h, GLA2h, GLA3h) levels were measured on 0 h, 0.5 h, 1 h, 2 h, and 3 h after a 100 g standard bread meal load. Early postprandial secretion function of islet β cells was calculated as Δcp0.5h = CP0.5-FCP. The patients were divided into low, medium and high groups (T1, T2 and T3) according to tertiles of OCN. Comparison of parameters among three groups was studied. Correlation analysis confirmed the relationship between OCN and pancreatic secretion. Multiple regression analysis showed independent contributors to pancreatic secretion. MAIN RESULTS FBG, and PBG2h were the lowest while Δcp0.5h was the highest in the highest tertile group (respectively, p < 0.05). INS3h, area under the curve of insulin (AUCins3h) in T3 Group were significantly lower than T1 Group (respectively, p < 0.05). GLA1h in T3 group was lower than T1 group (p < 0.05), and GLA0.5 h in T3 group was lower than T2 and T1 groups (p < 0.05). Correlation analysis showed OCN was inversely correlated with Homeostatic model of insulin resistance (HOMA-IR), INS3h, AUCins3h (p < 0.05), and was still inversely correlated with FCP, GLA0.5 h, GLA1h, area under the curve of glucagon (AUCgla3h) (respectively, p < 0.05) after adjustment for body mass index (BMI) and alanine aminotransferase (ALT). The multiple regression analysis showed that OCN was independent contributor to Δcp0.5h, GLA0.5h and GLA1h (respectively, p < 0.05). CONCLUSIONS Higher serum OCN level is closely related to better blood glucose control, higher insulin sensitivity, increased early-phase insulin secretion of islet β cells and appropriate inhibition of postprandial glucagon secretion of islet ɑ cells in adult patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Haiyan Lei
- Department of Endocrinology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Jun Liu
- Department of Endocrinology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Wei Wang
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China
| | - Xinyi Yang
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China
| | - Zhouqin Feng
- Department of Endocrinology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Pu Zang
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China
| | - Bin Lu
- Department of Endocrinology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China.
| | - Jiaqing Shao
- Department of Endocrinology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Shahrour HE, Al Fahom S, Al-Massarani G, AlSaadi AR, Magni P. Osteocalcin-expressing endothelial progenitor cells and serum osteocalcin forms are independent biomarkers of coronary atherosclerotic disease severity in male and female patients. J Endocrinol Invest 2022; 45:1173-1180. [PMID: 35089541 PMCID: PMC9098612 DOI: 10.1007/s40618-022-01744-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/14/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Osteocalcin (OC), an osteoblast-derived regulator of metabolic processes, and circulating early endothelial progenitor cells (EPC, CD34 - /CD133 + /KDR +) expressing OC (OC +) are potential candidates linking bone metabolism and the vasculature and might be involved in vascular atherosclerotic calcification. This study aimed at assessing the association of circulating levels of different OC forms and of EPCs count with disease severity in patients with documented coronary atherosclerosis (CAD). METHODS Patients (n = 59) undergoing coronary angiography were divided, according to stenosis severity, into (1) early coronary atherosclerosis (ECA) (n = 22), and (2) late coronary atherosclerosis (LCA) (n = 37). Total OC (TOC), carboxylated OC (cOC), undercarboxylated OC (unOC) were quantified by ELISA. EPC OC + count was assessed by flow cytometry. RESULTS EPC OC + counts showed significant differences between ECA and LCA groups. unOC and unOC/TOC ratio were inversely correlated with EPC OC + count. A significant decrease in TOC and unOC plasma levels was associated with higher cardiovascular risk factors (CVRFs) number. EPC OC + count was correlated with LDL-C, total cholesterol, and triglycerides, with a greater significance in the LCA group. No association between the different forms of circulating OC (TOC, ucOC, cOC) and severity of CAD was found. CONCLUSION This study showed a significant association between EPCs (CD34 - /CD133 + /KDR + /OC +), CAD severity and CVRFs, suggesting an active role for EPC OC + in the development of CAD. An inverse correlation between TOC, ucOC, and number of CVRFs was observed, suggesting that OC, regardless of its carboxylation status, may be developed as a further cardiovascular risk biomarker.
Collapse
Affiliation(s)
- H E Shahrour
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syria
| | - S Al Fahom
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syria
| | - G Al-Massarani
- Department Radiation Medicine, Pharmacological Studies Division, Atomic Energy Commission of Syria (AECS), Damascus, Syria
| | - A R AlSaadi
- Department of Internal Medicine, Cardiovascular Disease Section, Faculty of Medicine, Damascus University, Damascus, Syria
| | - P Magni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milan, Italy.
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy.
- DISFeB-UNIMI, via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
11
|
Xing Y, Liu J, Liu H, Ma H. Relationship of Bone Turnover Markers with Serum Uric Acid-to-Creatinine Ratio in Men and Postmenopausal Women with Type 2 Diabetes. Diabetes Metab Syndr Obes 2022; 15:3205-3217. [PMID: 36268198 PMCID: PMC9578772 DOI: 10.2147/dmso.s384694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Accumulating evidence has shown that serum uric acid (UA) is associated with some chronic diseases owing to its antioxidant capacity; however, previous research has discrepant results regarding the relationship between UA and bone health. UA normalized by renal function can reflect endogenous UA levels more precisely than SUA levels. This study assessed the relationship between serum UA-to-creatinine (SUA/Cr) ratio and bone turnover markers (BTMs) in men and postmenopausal women with type 2 diabetes mellitus (T2DM). PATIENTS AND METHODS Overall, 1691 patients (1028 males and 663 postmenopausal females) with T2DM admitted to Hebei General Hospital between January and December 2020 were selected and divided into two groups according to their SUA/Cr ratio. One-way analysis of variance was used to compare groups. The relationship between the SUA/Cr ratio and BTMs (including osteocalcin [OC], procollagen I N-terminal peptide [PINP], and β-isomerized type I collagen C-telopeptide breakdown products [β-CTX]) was analyzed using multiple linear regression. Furthermore, subgroup analyses were performed to explore the differences between men and women in the relationship between SUA/Cr and BTMs. Mediation analysis was used to explore whether insulin resistance mediated the association between SUA/Cr and BTMs. RESULTS β-CTX and PNIP levels of patients with T2DM in the low SUA/Cr group were significantly higher than those in the high SUA/Cr group, and the difference between the two groups was statistically significant (P < 0.05). Correlation analysis showed that SUA/Cr was negatively correlated with β-CTX and PNIP. After adjusting for confounding factors, multivariate linear regression analysis revealed that the SUA/Cr level was negatively correlated with PINP and β-CTX in male patients and postmenopausal women with T2DM. Stronger correlations were found in patients with 25(OH)D3 < 20ng/mL, course ≥ 5 years, HbA1c > 7%, or BMI < 28 kg/m2. CONCLUSION SUA/Cr ratio was an independent influencing factor of BTMs in patients with T2DM.
Collapse
Affiliation(s)
- Yuling Xing
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Jing Liu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Huanxin Liu
- Health Examination Center, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Huijuan Ma
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, People’s Republic of China
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, People’s Republic of China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Correspondence: Huijuan Ma, Department of Endocrinology, Hebei General Hospital, Shijiazhuang, People’s Republic of China, Tel +86 18032838686, Email
| |
Collapse
|
12
|
Gong Y, Li N, Lai M, Fang F, Yang J, Kang M, Shen T, Peng Y, Wang Y. Consistently Low Levels of Osteocalcin From Late Pregnancy to Postpartum Are Related to Postpartum Abnormal Glucose Metabolism in GDM Patients. Front Endocrinol (Lausanne) 2022; 13:803624. [PMID: 35321337 PMCID: PMC8936089 DOI: 10.3389/fendo.2022.803624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Increasing evidence suggests that osteocalcin (OC), a marker of bone formation, plays an important role in glucose homoeostasis. Few studies have investigated the relationship between OC levels in gestational diabetes mellitus (GDM) patients and their postpartum glucose metabolism. This study evaluated the relationship between OC levels in late pregnancy, their longitudinal changes, and postpartum glucose metabolism among GDM patients. MEASURES Serum OC was measured in late pregnancy and the postpartum period for 721 GDM patients. All patients underwent a 75-g oral glucose tolerance test (OGTT) at 6-8 weeks postpartum. According to postpartum OGTT outcomes, patients were categorized into abnormal glucose metabolism (AGM) (n=255) and normal glucose tolerance (NGT) groups (n=466). Glucose metabolism-related indices were measured and calculated. Logistic regression analysis and linear mixed-effects model were used to assess the association between OC and postpartum AGM. RESULTS In late pregnancy, OC levels were lower in the AGM group than in the NGT group (13.93 ± 6.90 vs 15.33 ± 7.63 ng/ml, P=0.015). After delivery, OC levels increased in both groups. However, OC levels remained lower in the AGM group than in the NGT group (23.48 ± 7.84 vs 25.65 ± 8.37 ng/ml, P=0.001). Higher OC levels in late pregnancy were associated with decreased risk of progressing to postpartum AGM (OR:0.96, 95%CI:0.94-0.99). Linear mixed-effects analysis showed that postpartum AGM patients exhibited consistently lower OC levels than NGT group from late pregnancy to the postpartum period after adjustment for cofactors (β=-1.70, 95% CI: -2.78- -0.62). CONCLUSIONS In GDM patients, consistently low levels of OC from late pregnancy to postpartum were associated with increased postpartum AGM risk. The increase in serum OC may act as a protective factor to curb the progression of AGM at postpartum for GDM patients.
Collapse
Affiliation(s)
- Yujia Gong
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Na Li
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyu Lai
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Fang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaying Yang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mei Kang
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Shen
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongde Peng
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufan Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yufan Wang,
| |
Collapse
|
13
|
Schilling K, Brown E, Zhang X. NAD(P)H autofluorescence lifetime imaging enables single cell analyses of cellular metabolism of osteoblasts in vitro and in vivo via two-photon microscopy. Bone 2022; 154:116257. [PMID: 34781049 PMCID: PMC8671374 DOI: 10.1016/j.bone.2021.116257] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023]
Abstract
Two-photon fluorescence lifetime microscopy (2P-FLIM) is a non-invasive optical technique that can obtain cellular metabolism information based on the intrinsic autofluorescence lifetimes of free and enzyme-bound NAD(P)H, which reflect the metabolic state of single cells within the native microenvironment of the living tissue. NAD(P)H 2P-FLIM was initially performed in bone marrow stromal cell (BMSC) cultures established from Col (I) 2.3GFP or OSX-mCherry mouse models, in which osteoblastic lineage cells were labelled with green or red fluorescence protein, respectively. Measurement of the mean NAD(P)H lifetime, τM, demonstrated that osteoblasts in osteogenic media had a progressively increased τM compared to cells in regular media, suggesting that osteoblasts undergoing mineralization had higher NAD+/NAD(P)H ratio and may utilize more oxidative phosphorylation (OxPhos). In vivo NAD(P)H 2P-FLIM was conducted in conjunction with two-photon phosphorescence lifetime microscopy (2P-PLIM) to evaluate cellular metabolism of GFP+ osteoblasts as well as bone tissue oxygen at different locations of the native cranial bone in Col (I) 2.3GFP mice. Our data showed that osteocytes dwelling within lacunae had higher τM than osteoblasts at the bone edge of suture and marrow space. Measurement of pO2 showed poor correlation of pO2 and τM in native bone. However, when NAD(P)H 2P-FLIM was used to examine osteoblast cellular metabolism at the leading edge of the cranial defects during repair in Col (I) 2.3GFP mouse model, a significantly lower τM was recorded, which was associated with lower pO2 at an early stage of healing, indicating an impact of hypoxia on energy metabolism during bone tissue repair. Taken together, our current study demonstrates the feasibility of using non-invasive optical NAD(P)H 2P-FLIM technique to examine cellular energy metabolism at single cell resolution in living animals. Our data further support that both glycolysis and OxPhos are being used in the osteoblasts, with more mature osteoblasts exhibiting higher ratio of NAD+/NAD(P)H, indicating a potential change of energy mode during differentiation. Further experiments utilizing animals with genetic modification of cellular metabolism could enhance our understanding of energy metabolism in various cell types in living bone microenvironment.
Collapse
Affiliation(s)
- Kevin Schilling
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Edward Brown
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
14
|
Khosla S, Samakkarnthai P, Monroe DG, Farr JN. Update on the pathogenesis and treatment of skeletal fragility in type 2 diabetes mellitus. Nat Rev Endocrinol 2021; 17:685-697. [PMID: 34518671 PMCID: PMC8605611 DOI: 10.1038/s41574-021-00555-5] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 02/08/2023]
Abstract
Fracture risk is increased in patients with type 2 diabetes mellitus (T2DM). In addition, these patients sustain fractures despite having higher levels of areal bone mineral density, as measured by dual-energy X-ray absorptiometry, than individuals without T2DM. Thus, additional factors such as alterations in bone quality could have important roles in mediating skeletal fragility in patients with T2DM. Although the pathogenesis of increased fracture risk in T2DM is multifactorial, impairments in bone material properties and increases in cortical porosity have emerged as two key skeletal abnormalities that contribute to skeletal fragility in patients with T2DM. In addition, indices of bone formation are uniformly reduced in patients with T2DM, with evidence from mouse studies published over the past few years linking this abnormality to accelerated skeletal ageing, specifically cellular senescence. In this Review, we highlight the latest advances in our understanding of the mechanisms of skeletal fragility in patients with T2DM and suggest potential novel therapeutic approaches to address this problem.
Collapse
Affiliation(s)
- Sundeep Khosla
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Parinya Samakkarnthai
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Division of Endocrinology, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - David G Monroe
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Joshua N Farr
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
15
|
Suchacki KJ, Alcaide-Corral CJ, Nimale S, Macaskill MG, Stimson RH, Farquharson C, Freeman TC, Tavares AAS. A Systems-Level Analysis of Total-Body PET Data Reveals Complex Skeletal Metabolism Networks in vivo. Front Med (Lausanne) 2021; 8:740615. [PMID: 34616758 PMCID: PMC8488174 DOI: 10.3389/fmed.2021.740615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Bone is now regarded to be a key regulator of a number of metabolic processes, in addition to the regulation of mineral metabolism. However, our understanding of complex bone metabolic interactions at a systems level remains rudimentary. in vitro molecular biology and bioinformatics approaches have frequently been used to understand the mechanistic changes underlying disease at the cell level, however, these approaches lack the capability to interrogate dynamic multi-bone metabolic interactions in vivo. Here we present a novel and integrative approach to understand complex bone metabolic interactions in vivo using total-body positron emission tomography (PET) network analysis of murine 18F-FDG scans, as a biomarker of glucose metabolism in bones. In this report we show that different bones within the skeleton have a unique glucose metabolism and form a complex metabolic network, which could not be identified using single tissue simplistic PET standard uptake values analysis. The application of our approach could reveal new physiological and pathological tissue interactions beyond skeletal metabolism, due to PET radiotracers diversity and the advent of clinical total-body PET systems.
Collapse
Affiliation(s)
- Karla J. Suchacki
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos J. Alcaide-Corral
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | - Samah Nimale
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark G. Macaskill
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | - Roland H. Stimson
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Colin Farquharson
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies (RDSVS), Easter Bush Campus, University of Edinburgh, Edinburgh, United Kingdom
| | - Tom C. Freeman
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies (RDSVS), Easter Bush Campus, University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A. S. Tavares
- University/British Heart Foundation (BHF) Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Behl T, Sehgal A, Grover M, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Aleya L, Bungau S. Uncurtaining the pivotal role of ABC transporters in diabetes mellitus. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:41533-41551. [PMID: 34085197 DOI: 10.1007/s11356-021-14675-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/27/2021] [Indexed: 06/12/2023]
Abstract
The metabolic disorders are the edge points for the initiation of various diseases. These disorders comprised of several diseases including diabetes, obesity, and cardiovascular complications. Worldwide, the prevalence of these disorders is increasing day by day. The world's population is at higher threat of developing metabolic disease, especially diabetes. Therefore, there is an impregnable necessity of searching for a newer therapeutic target to reduce the burden of these disorders. Diabetes mellitus (DM) is marked with the dysregulated insulin secretion and resistance. The lipid and glucose transporters portray a pivotal role in the metabolism and transport of both of these. The excess production of lipid and glucose and decreased clearance of these leads to the emergence of DM. The ATP-binding cassette transporters (ABCT) are important for the metabolism of glucose and lipid. Various studies suggest the key involvement of ABCT in the pathologic process of different diseases. In addition, the involvement of other pathways, including IGF signaling, P13-Akt/PKC/MAPK signaling, and GLP-1 via regulation of ABCT, may help develop new treatment strategies to cope with insulin resistance dysregulated glucose metabolism, key features in DM.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Madhuri Grover
- BS Anangpuria Institute of Pharmacy, Faridabad, Haryana, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University, Gurugram, Haryana, India
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Simona Bungau
- Department of Pharmacy, Faculty of Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
17
|
Whelan IT, Moeendarbary E, Hoey DA, Kelly DJ. Biofabrication of vasculature in microphysiological models of bone. Biofabrication 2021; 13. [PMID: 34034238 DOI: 10.1088/1758-5090/ac04f7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 05/25/2021] [Indexed: 11/12/2022]
Abstract
Bone contains a dense network of blood vessels that are essential to its homoeostasis, endocrine function, mineral metabolism and regenerative functions. In addition, bone vasculature is implicated in a number of prominent skeletal diseases, and bone has high affinity for metastatic cancers. Despite vasculature being an integral part of bone physiology and pathophysiology, it is often ignored or oversimplified inin vitrobone models. However, 3D physiologically relevant vasculature can now be engineeredin vitro, with microphysiological systems (MPS) increasingly being used as platforms for engineering this physiologically relevant vasculature. In recent years, vascularised models of bone in MPSs systems have been reported in the literature, representing the beginning of a possible technological step change in how bone is modelledin vitro. Vascularised bone MPSs is a subfield of bone research in its nascency, however given the impact of MPSs has had inin vitroorgan modelling, and the crucial role of vasculature to bone physiology, these systems stand to have a substantial impact on bone research. However, engineering vasculature within the specific design restraints of the bone niche is significantly challenging given the different requirements for engineering bone and vasculature. With this in mind, this paper aims to serve as technical guidance for the biofabrication of vascularised bone tissue within MPS devices. We first discuss the key engineering and biological considerations for engineering more physiologically relevant vasculaturein vitrowithin the specific design constraints of the bone niche. We next explore emerging applications of vascularised bone MPSs, and conclude with a discussion on the current status of vascularised bone MPS biofabrication and suggest directions for development of next generation vascularised bone MPSs.
Collapse
|
18
|
Disentangling the relationship between bone turnover and glucose homeostasis: A prospective, population-based twin study. Bone Rep 2021; 14:100752. [PMID: 33665235 PMCID: PMC7900018 DOI: 10.1016/j.bonr.2021.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/02/2021] [Indexed: 11/23/2022] Open
Abstract
Background Biochemical markers of bone turnover are lower in patients with type 2 diabetes, which may be explained by genetic variants being associated with type 2 diabetes and bone turnover as well as environmental factors. We hypothesized that bone turnover markers associate with and predict changes in glucose homeostasis after control for genetics and shared environment. Methods 1071 healthy, non-diabetic (at baseline, 1997-2000) adult mono- and dizygotic twins participating in the prospective study GEMINAKAR were reassessed between 2010 and 2012 with clinical evaluation, biochemical tests and oral glucose tolerance test. Fasting bone turnover markers (CTX, P1NP and osteocalcin) were measured. The association between bone turnover, glucose homeostasis and the ability of bone turnover markers to predict changes in glucose homeostasis were assessed in cross-sectional and longitudinal analyses. Analyses were performed both at an individual level and adjusted for shared environmental and genetic factors. Results Glucose levels increased with age, and 33 (3%) participants had developed type 2 diabetes at follow-up. In women, bone turnover markers increased with age, whereas for men only osteocalcin increased with age. Bone turnover markers were not associated with fasting glucose, insulin, or HOMA-IR at baseline or follow-up before or after adjustment for age, sex, BMI, smoking, and use of medication at baseline. Variation in bone turnover markers was mainly explained by unique environmental factors, 70%, 70% and 55% for CTX, P1NP and osteocalcin, respectively, whereas additive genetic factors explained 7%, 13% and 45% of the variation in CTX, P1NP and osteocalcin. Conclusions Bone turnover markers were not associated with baseline plasma glucose levels and did not predict changes in glucose homeostasis. Variation in bone turnover markers is mainly explained by environmental factors, however, compared to CTX and P1NP, genetic factors have a larger impact on osteocalcin levels.
Collapse
|
19
|
Moon JS, Jin MH, Koh HM. Association between Serum Osteocalcin Levels and Metabolic Syndrome according to the Menopausal Status of Korean Women. J Korean Med Sci 2021; 36:e56. [PMID: 33650335 PMCID: PMC7921371 DOI: 10.3346/jkms.2021.36.e56] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/16/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Osteocalcin is known to regulate energy metabolism. Recently, metabolic syndrome (MetS) has been found to be associated with reduced levels of osteocalcin in men, as well as in postmenopausal women. The aim of this study was to investigate the association between serum osteocalcin and MetS in premenopausal women, compared with that in postmenopausal women. METHODS This cross-sectional study was based on 5,896 participants who completed a health screening examination. They were classified according to their menopausal status. Each group was subdivided into non-MetS and MetS groups according to the modified National Cholesterol Education Program-Adult Treatment Panel III criteria. Serum osteocalcin levels were measured using the electrochemiluminescence immunoassay. RESULTS Serum osteocalcin level was significantly lower in women with MetS than in those without MetS, after adjusting for confounders (14.12 ± 0.04 vs. 13.17 ± 0.13 [P = 0.004] in premenopausal women, and 20.34 ± 0.09 vs. 19.62 ± 0.21 [P < 0.001] in postmenopausal women), regardless of their menopausal status. Serum osteocalcin levels decreased correspondingly with an increasing number of MetS elements (P for trend < 0.001). Multiple regression analysis demonstrated that waist circumference (β = -0.085 [P < 0.001] and β = -0.137 [P < 0.001]) and hemoglobin A1c (β = -0.09 [P < 0.001] and β = -0.145 [P < 0.001]) were independent predictors of osteocalcin in premenopausal and postmenopausal women. Triglyceride levels were also independently associated with osteocalcin levels in premenopausal women (β = -0.004 [P < 0.013]). The odds ratio (OR) for MetS was significantly higher in the lowest quartile than in the highest quartile of serum osteocalcin levels after adjusting for age, alkaline phosphatase, uric acid, high sensitivity C-reactive protein, and body mass index in all women (OR, 2.00; 95% confidence interval [CI], 1.49-2.68) as well as in premenopausal (OR, 2.23; 95% CI, 1.39-3.58) and postmenopausal (OR, 2.01; 95% CI, 1.26-3.23) subgroups. CONCLUSION Lower serum osteocalcin concentrations were significantly associated with MetS in both premenopausal and postmenopausal women and were therefore independent of menopausal status.
Collapse
Affiliation(s)
- Jin Sook Moon
- Department of Family Medicine, Samsung Changwon Hospital, School of Medicine, Sungkyunkwan University, Changwon, Korea
- Department of Family Medicine, Pusan National University Graduate School of Medicine, Busan, Korea
| | - Mi Hyeon Jin
- Department of Research Support, Samsung Changwon Hospital, School of Medicine, Sungkyunkwan University, Changwon, Korea
| | - Hyun Min Koh
- Department of Family Medicine, Samsung Changwon Hospital, School of Medicine, Sungkyunkwan University, Changwon, Korea.
| |
Collapse
|
20
|
An Y, Liu S, Wang W, Dong H, Zhao W, Ke J, Zhao D. Low serum levels of bone turnover markers are associated with the presence and severity of diabetic retinopathy in patients with type 2 diabetes mellitus. J Diabetes 2021; 13:111-123. [PMID: 32671958 DOI: 10.1111/1753-0407.13089] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/18/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Accumulating evidence demonstrates an association of type 2 diabetes mellitus (T2DM) and its microvascular complications with increased fracture risk. In this study, we aimed to evaluate the relationships between serum concentrations of bone turnover markers and the presence and/or severity of diabetic retinopathy (DR) among patients with T2DM. METHODS A total of 285 patients with T2DM comprising 168 patients without DR and 117 patients with DR were enrolled in the cross-sectional study. In the latter group, patients were further divided into patients of mild and severe DR stages. The biochemical parameters and bone turnover markers were determined in all participants. RESULTS This study found that serum levels of procollagen type 1 N-terminal propeptide (P1NP), a bone formation marker, and the bone resorption marker serum β-cross-linked C-telopeptide of type I collagen (β-CTX) were more decreased in diabetic patients with DR than in those without DR, with differences remaining significant (P < .05) in multivariate linear regression models after adjustments for multiple confounding factors. Osteocalcin and β-CTX levels were further reduced along with the severity of DR among participants with DR. Moreover, multivariate logistic regression analysis revealed that lower serum levels of P1NP and β-CTX were associated with higher odds for the presence of DR, while β-CTX was associated with the severity of DR. CONCLUSION Our results suggest that the development of DR might be involved in the progression of T2DM-induced deficits in bone formation and resorption or vice versa. Follow-up studies and further research are necessary to validate the associations and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yaxin An
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Simo Liu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Wenbo Wang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Huan Dong
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Wenying Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| |
Collapse
|
21
|
Desentis-Desentis MF, Rivas-Carrillo JD, Sánchez-Enríquez S. Protective role of osteocalcin in diabetes pathogenesis. J Bone Miner Metab 2020; 38:765-771. [PMID: 32725267 DOI: 10.1007/s00774-020-01130-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/08/2020] [Indexed: 12/18/2022]
Abstract
In diabetes, metabolic, inflammatory, and stress-associated alterations conduce to ß-cell failure and tissue damage. Osteocalcin is a bone protein with several endocrine functions in different tissues. In this review, we gathered scientific evidence of how osteocalcin could modulate functional disorders that are altered in diabetes in an integrative way. We include adipose tissue, pancreatic function, and oxidative stress aspects. In the first section, we focus on the role of inflammatory mediators and adiponectin in energy homeostasis and insulin sensitivity. In the following section, we discuss the effect of osteocalcin in metabolic and pancreatic function and its association in insulin signaling and in ß-cell proliferation. Finally, we focus on osteocalcin action in oxidative and endoplasmic reticulum stress, and in antioxidant regulation, since ß-cells are well known by its vulnerability to stress damage. These evidences support the notion that osteocalcin could have an important role in diabetes treatment.
Collapse
Affiliation(s)
- María Fernanda Desentis-Desentis
- Laboratory of Tissue Engineering and Transplant, Department of Physiology, University Center for Health Sciences, University de Guadalajara, 950 Sierra Mojada St., Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
- cGMP Cell Processing Facility, University Center for Health Sciences, University of Guadalajara, 950 Sierra Mojada St., Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - Jorge David Rivas-Carrillo
- Laboratory of Tissue Engineering and Transplant, Department of Physiology, University Center for Health Sciences, University de Guadalajara, 950 Sierra Mojada St., Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
- cGMP Cell Processing Facility, University Center for Health Sciences, University of Guadalajara, 950 Sierra Mojada St., Col. Independencia, C.P. 44340, Guadalajara, Jalisco, Mexico
| | - Sergio Sánchez-Enríquez
- Department of Clinics, University Center of Los Altos, University of Guadalajara, 1200 Rafael Casillas Ave, ZC47620, Tepatitlán de Morelos, Jalisco, Mexico.
| |
Collapse
|
22
|
Ducy P. Bone Regulation of Insulin Secretion and Glucose Homeostasis. Endocrinology 2020; 161:5895464. [PMID: 32822470 DOI: 10.1210/endocr/bqaa149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/18/2020] [Indexed: 12/31/2022]
Abstract
For centuries our image of the skeleton has been one of an inert structure playing a supporting role for muscles and a protective role for inner organs like the brain. Cell biology and physiology modified this view in the 20st century by defining the constant interplay between bone-forming and bone resorbing cells that take place during bone growth and remodeling, therefore demonstrating that bone is as alive as any other tissues in the body. During the past 40 years human and, most important, mouse genetics, have allowed not only the refinement of this notion by identifying the many genes and regulatory networks responsible for the crosstalk existing between bone cells, but have redefined the role of bone by showing that its influence goes way beyond its own physiology. Among its newly identified functions is the regulation of energy metabolism by 2 bone-derived hormones, osteocalcin and lipocalin-2. Their biology and respective roles in this process are the topic of this review.
Collapse
Affiliation(s)
- Patricia Ducy
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, College of Physicians and Surgeons, New York, New York
| |
Collapse
|
23
|
Shtaif B, Bar-Maisels M, Gabet Y, Hiram-Bab S, Yackobovitch-Gavan M, Phillip M, Gat-Yablonski G. Cartilage -specific knockout of Sirt1 significantly reduces bone quality and catch-up growth efficiency. Bone 2020; 138:115468. [PMID: 32512163 DOI: 10.1016/j.bone.2020.115468] [Citation(s) in RCA: 10] [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: 03/05/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Spontaneous catch-up (CU) growth occurs when a growth-restricting factor is resolved. However, its efficiency is sometimes inadequate and growth deficits remain permanent. The therapeutic toolbox for short stature is currently very limited, thus, finding new regulatory pathways is important for the development of novel means of treatment. Our previous studies using a nutrition-induced CU growth model showed that the level of sirtuin-1 (Sirt1) was significantly increased in food-restricted animals and decreased during CU growth. AIM This study sought to investigate the role of Sirt1 in modulating the response of the epiphyseal growth plate (EGP) to nutritional manipulation. METHOD Collagen type II-specific Sirt1 knockout (CKO) mice were tested for response to our CU growth model consisting of a period of food restriction followed by re-feeding. RESULTS The transgenic CKO mice weighed more than the control (CTL) mice, their EGP was higher and less organized, specifically at the resting and proliferative zones, leading to shorter bones. Ablation of Sirt1 in the chondrocytes was found to have a dramatic effect on bone mineralization on micro-CT analysis. The CKO mice were less responsive to the nutritional manipulation, and their CU growth was less efficient. They remained shorter than the CTL mice who corrected the food restriction-induced growth deficit during the re-feeding period. CONCLUSIONS Sirt1 appears to be important for normal regulation of the EGP. In its absence, the EGP is less organized and CU growth is less efficient. These results suggest that SIRT1 may serve as a novel therapeutic target for short stature.
Collapse
Affiliation(s)
- Biana Shtaif
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Felsenstein Medical Research Center, Petach Tikva, Israel.
| | - Meytal Bar-Maisels
- Felsenstein Medical Research Center, Petach Tikva, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Yankel Gabet
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Sahar Hiram-Bab
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Michal Yackobovitch-Gavan
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Moshe Phillip
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Felsenstein Medical Research Center, Petach Tikva, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | - Galia Gat-Yablonski
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Felsenstein Medical Research Center, Petach Tikva, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| |
Collapse
|
24
|
Affiliation(s)
- Ravinder Goswami
- Department of Endocrinology & Metabolism, All India Institute of Medical Sciences, New Delhi 110 029, India
| | - Abilash Nair
- Department of Endocrinology & Metabolism, Government Medical College, Thiruvananthapuram 695 011, Kerala, India
| |
Collapse
|
25
|
Zhu B, Lin Z, Chen X, Gusdon AM, Shen W, Chen J, Zheng L, Sun H, Li Y, Zhu C, Li J, Qu S. Circulating osteocalcin: A potential predictor of ketosis in type 2 diabetes. Diabetes Metab Res Rev 2020; 36:e3265. [PMID: 31850672 DOI: 10.1002/dmrr.3265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 07/05/2019] [Accepted: 12/11/2019] [Indexed: 12/28/2022]
Abstract
AIMS Osteocalcin contributes to the regulation of endocrine system. However, the association between osteocalcin and ketosis has not been evaluated. We thus aimed to explore the relationship between total osteocalcin and risk of ketosis in type 2 diabetes (T2DM). MATERIALS AND METHODS We identified 6157 diabetes patients from Shanghai Tenth People's Hospital between 1 January 2011 and 1 March 2017. Six hundred eight subjects were enrolled in the retrospective cross-sectional study: 304 T2DM patients with ketosis whose age, gender, and body mass index were matched with 304 T2DM patients without ketosis. A further retrospective nested case-control study was conducted in 252 T2DM patients without ketosis for a mean duration of 21.58 ± 12.43 months to investigate the occurrence of ketosis. RESULTS Osteocalcin levels were negatively correlated with blood ketones (adjusted r = -0.263) and urine ketones (adjusted r = -0.183). The inverse dose-dependent relationship of osteocalcin and risk of ketosis was present across osteocalcin level quintiles (top quintile as the reference, adjusted odds ratio [95% CI] = 2.56 [0.80-8.17], 3.71 [0.90-15.29], 10.77 [2.63-44.15], 23.81 [4.32-131.17] per osteocalcin quintile, respectively). Ketosis occurred in 17 of the 252 T2DM patients during follow-up. The Cox regression analysis indicated that osteocalcin was an independent protective factor against development of ketosis (adjusted hazard ratio [95% CI]: 0.668 [0.460-0.971]). CONCLUSIONS Total osteocalcin can be used as a predictor of ketosis in T2DM.
Collapse
Affiliation(s)
- Bing Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- National Metabolic Management Center, Shanghai, China
| | - Ziwei Lin
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- National Metabolic Management Center, Shanghai, China
| | - Xianying Chen
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Endocrinology, School of Medicine, Nanjing Medical University, Nanjing, China
| | - Aaron M Gusdon
- Division of Neurocritical Care, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Wenjun Shen
- Division of Endocrinology,Gerontology and Metabolism, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo AltoHealth Care System, Palo Alto, CA, USA
| | - Jia Chen
- Department of Endocrinology, Beijing Jishuitan Hospital, 4th Medical College of Peking University, Beijing, China
| | - Liang Zheng
- Heart, Lung and Blood Vessel Center, Tongji University, Shanghai, China
| | - Hang Sun
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- National Metabolic Management Center, Shanghai, China
| | - Yan Li
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- National Metabolic Management Center, Shanghai, China
| | - Cuiling Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- National Metabolic Management Center, Shanghai, China
| | - Jue Li
- Heart, Lung and Blood Vessel Center, Tongji University, Shanghai, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- National Metabolic Management Center, Shanghai, China
- Department of Endocrinology, School of Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Shahen VA, Gerbaix M, Koeppenkastrop S, Lim SF, McFarlane KE, Nguyen ANL, Peng XY, Weiss NB, Brennan-Speranza TC. Multifactorial effects of hyperglycaemia, hyperinsulinemia and inflammation on bone remodelling in type 2 diabetes mellitus. Cytokine Growth Factor Rev 2020; 55:109-118. [PMID: 32354674 DOI: 10.1016/j.cytogfr.2020.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 04/08/2020] [Indexed: 12/14/2022]
Abstract
Bones undergo continuous cycles of bone remodelling that rely on the balance between bone formation and resorption. This balance allows the bone to adapt to changes in mechanical loads and repair microdamages. However, this balance is susceptible to upset in various conditions, leading to impaired bone remodelling and abnormal bones. This is usually indicated by abnormal bone mineral density (BMD), an indicator of bone strength. Despite this, patients with type 2 diabetes mellitus (T2DM) exhibit normal to high BMD, yet still suffer from an increased risk of fractures. The activity of the bone cells is also altered as indicated by the reduced levels of bone turnover markers in T2DM observed in the circulation. The underlying mechanisms behind these skeletal outcomes in patients with T2DM remain unclear. This review summarises recent findings regarding inflammatory cytokine factors associated with T2DM to understand the mechanisms involved and considers potential therapeutic interventions.
Collapse
Affiliation(s)
- V A Shahen
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
| | - M Gerbaix
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital & Faculty of Medicine, Geneva, Switzerland
| | - S Koeppenkastrop
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
| | - S F Lim
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
| | - K E McFarlane
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Amanda N L Nguyen
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
| | - X Y Peng
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
| | - N B Weiss
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
| | - T C Brennan-Speranza
- Department of Physiology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia; School of Public Health, Faculty of Medicine and Health, The University of Sydney, Australia.
| |
Collapse
|
27
|
Rendina-Ruedy E, Rosen CJ. Lipids in the Bone Marrow: An Evolving Perspective. Cell Metab 2020; 31:219-231. [PMID: 31668874 PMCID: PMC7004849 DOI: 10.1016/j.cmet.2019.09.015] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/05/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
Because of heavy energy demands to maintain bone homeostasis, the skeletal system is closely tied to whole-body metabolism via neuronal and hormonal mediators. Glucose, amino acids, and fatty acids are the chief fuel sources for bone resident cells during its remodeling. Lipids, which can be mobilized from intracellular depots in the bone marrow, can be a potent source of fatty acids. Thus, while it has been suggested that adipocytes in the bone marrow act as "filler" and are detrimental to skeletal homeostasis, we propose that marrow lipids are, in fact, essential for proper bone functioning. As such, we examine the prevailing evidence regarding the storage, use, and export of lipids within the skeletal niche, including from both in vitro and in vivo model systems. We also highlight the numerous challenges that remain to fully appreciate the relationship of lipid turnover to skeletal homeostasis.
Collapse
Affiliation(s)
- Elizabeth Rendina-Ruedy
- Center for Molecular Medicine, Research Institute, Maine Medical Center, Scarborough, ME 04074, USA; Vanderbilt Center for Bone Biology, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Clifford J Rosen
- Center for Molecular Medicine, Research Institute, Maine Medical Center, Scarborough, ME 04074, USA
| |
Collapse
|
28
|
Lewis KT, MacDougald OA. Local interactions in the bone marrow microenvironment and their contributions to systemic metabolic processes. LIPID SIGNALING AND METABOLISM 2020:63-80. [DOI: 10.1016/b978-0-12-819404-1.00004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
29
|
Funakoshi S, Yoshimura K, Hirano S, Ohmi S, Amano E, Fukuda Y, Terada Y, Fujimoto S. Undercarboxylated osteocalcin correlates with insulin secretion in Japanese individuals with diabetes. Diabetol Metab Syndr 2020; 12:72. [PMID: 32821293 PMCID: PMC7433182 DOI: 10.1186/s13098-020-00579-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/12/2020] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Undercarboxylated osteocalcin (ucOC) is a secreted protein produced by osteoblasts that regulates insulin secretion and insulin sensitivity in rodents. However, the significance of these effects on glucose metabolism in human remains unknown. Moreover, the pathophysiological roles of ucOC on varying degrees of glucose intolerance, including diabetes need to be elucidated. In the present study, correlations between ucOC and indices of insulin secretion and sensitivity were analyzed in normal glucose tolerance (NGT), impaired glucose metabolism (IGM), and diabetes mellitus (DM) groups. METHODS Based on 75 g OGTT data in Japanese individuals without diabetic medication, or medications which may affect ucOC levels, individuals were classified as having normal glucose tolerance (NGT), impaired glucose metabolism (IGM), or diabetes (DM). In each group, 25 individuals were consecutively recruited [total 75 individuals, age: 65 ± 11 (mean ± SD); BMI: 24.9 ± 3.8 kg/m2]. QUICKI and Matsuda index (MI) were calculated as insulin sensitivity indices. Homeostasis model assessment (HOMA)-β and insulinogenic index (IGI) were calculated as insulin secretion indices. UcOC was measured using ECLIA. Normally-distributed loge-transformed (ln-) values were used for ucOC, HOMA-β, IGI, and MI. RESULTS The ucOC was not significantly different among the three groups. The results of multiple regression analysis showed that ln-ucOC did not significantly correlate with age, sex, BMI, waist circumference, fasting plasma glucose, plasma glucose 120 min after glucose loading, fasting plasma immunoreactive insulin, ln-HOMA-β, QUICKI, or ln-MI in any of the three groups. Interestingly, ln-ucOC correlated with ln-IGI (r = 0.422, P = 0.0354) and HbA1c (r = - 0.574, P = 0.0027) only in the DM group. There was no significant correlation between ln-IGI and age, sex, BMI, or HbA1c in the DM group. Further, the results of multiple regression analysis showed that ln-IGI could be independently predicted by BMI (β = 0.598, P = 0.0014) and ln-ucOC (β = 0.641, P = 0.0007) in the DM group (R2 = 0.488, P = 0.0006). CONCLUSION In our study, ucOC positively correlated with insulin secretion independently of BMI in Japanese individuals with diabetes. These results suggest that ucOC plays more important roles in insulin secretion than in insulin sensitivity in individuals with diabetes.
Collapse
Affiliation(s)
- Shogo Funakoshi
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505 Japan
| | - Kumiko Yoshimura
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505 Japan
- Fukuda Clinic, Kochi, Kochi 780-0023 Japan
| | - Seiki Hirano
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505 Japan
| | - Satoko Ohmi
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505 Japan
| | - Eri Amano
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505 Japan
| | | | - Yoshio Terada
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505 Japan
| | - Shimpei Fujimoto
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505 Japan
| |
Collapse
|
30
|
Yi Y, Du L, Qin M, Chen XQ, Sun XN, Li C, Du LJ, Liu Y, Liu Y, Sun JY, Tang Z, Xu M, Fang B, Liu X, Duan SZ. Regulation of Atrial Fibrosis by the Bone. Hypertension 2019; 73:379-389. [PMID: 30595118 DOI: 10.1161/hypertensionaha.118.11544] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
MR (mineralocorticoid receptor) antagonists have been demonstrated to provide beneficial effects on preventing atrial fibrosis. However, the underlying cellular and molecular mechanisms remain unclear. We aim to determine the role of osteoblast MR in atrial fibrosis and to explore the underlying mechanism. Using osteoblast MR knockout mouse in combination with mutant TGF (transforming growth factor)-β1 transgenic mouse, we demonstrated that MR deficiency in osteoblasts significantly attenuated atrial fibrosis. Mechanistically, MR directly regulated expression of OCN (osteocalcin) in osteoblasts. Both carboxylated and undercarboxylated OCNs (ucOC) were less secreted in osteoblast MR knockout mice. Mutant TGF-β1 transgenic mice supplemented with recombinant ucOC showed aggravated atrial fibrosis. In cultured atrial fibroblasts, ucOC treatment promoted proliferation and migration of atrial fibroblasts, whereas cotreatment with an antagonist for a GPRC6A (G-protein-coupled receptor, family C, group 6, member A) abolished these effects. Western blotting analysis revealed upregulation of PKA (protein kinase A) and CREB (cAMP-response element-binding protein) phosphorylation after ucOC treatment. Inhibition of PKA with its antagonist reduced ucOC-induced proliferation and migration of atrial fibroblasts. Finally, the impact of osteoblast MR deficiency on atrial fibrosis was abolished by ucOC administration in mutant TGF-β1 transgenic mice. Taken together, MR deficiency in osteoblasts attenuated atrial fibrosis by downregulation of OCN to promote proliferation and migration of atrial fibroblasts.
Collapse
Affiliation(s)
- Yi Yi
- From the Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, China (Y.Y., L.D., M.Q., X.-Q.C., X.L.)
| | - Lili Du
- From the Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, China (Y.Y., L.D., M.Q., X.-Q.C., X.L.)
| | - Mu Qin
- From the Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, China (Y.Y., L.D., M.Q., X.-Q.C., X.L.)
| | - Xiao-Qing Chen
- From the Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, China (Y.Y., L.D., M.Q., X.-Q.C., X.L.)
| | - Xue-Nan Sun
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences (X.-N.S., Yuan Liu).,Laboratory of Oral Microbiota and Systemic Diseases (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., S.-Z.D.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Chao Li
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas (C.L.)
| | - Lin-Juan Du
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Laboratory of Oral Microbiota and Systemic Diseases (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., S.-Z.D.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Yuan Liu
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences (X.-N.S., Yuan Liu).,Laboratory of Oral Microbiota and Systemic Diseases (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., S.-Z.D.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Yan Liu
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Laboratory of Oral Microbiota and Systemic Diseases (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., S.-Z.D.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Jian-Yong Sun
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Laboratory of Oral Microbiota and Systemic Diseases (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., S.-Z.D.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Zisheng Tang
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Department of Endodontics (Z.T.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Min Xu
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Department of Orthodontics (M.X., B.F.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Bing Fang
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Department of Orthodontics (M.X., B.F.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| | - Xu Liu
- From the Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, China (Y.Y., L.D., M.Q., X.-Q.C., X.L.)
| | - Sheng-Zhong Duan
- National Clinical Research Center for Oral Diseases, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, China (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., Z.T., M.X., B.F., S.-Z.D.).,Laboratory of Oral Microbiota and Systemic Diseases (X.-N.S., L.-J.D., Yuan Liu, Yan Liu, J.-Y.S., S.-Z.D.), Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
31
|
Yang L, Hu X, Zhang H, Pan W, Yu W, Gu X. Association of bone mineral density with a first-degree family history of diabetes in normoglycemic postmenopausal women. Menopause 2019; 26:1284-1288. [PMID: 31688576 DOI: 10.1097/gme.0000000000001396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE A first-degree family history of diabetes (FHD) contributes to increased risks of metabolic and cardiovascular diseases. Bone is an insulin-resistant site and an organ susceptible to microvascular complications. The goal of the present study was to investigate the association of FHD with bone mineral density (BMD) in postmenopausal women. METHODS In all, 892 normoglycemic postmenopausal women were divided into subgroups of participants with or without a first-degree FHD. BMD was measured using dual-energy x-ray absorptiometry. Fasting plasma insulin and glucose levels were measured, and insulin resistance was evaluated using the Homeostasis Model Assessment-Insulin Resistance (HOMA-IR) index. RESULTS The BMD of the lumbar spine and femoral neck were much higher in the participants with a first-degree FHD than in those without an FHD (all P < 0.05). Lumbar spine BMD and femoral neck BMD were both positively associated with HOMA-IR (P = 0.041 and P = 0.005, respectively). Multiple stepwise regression analysis showed that a first-degree FHD was an independent factor that was positively associated with lumbar spine BMD (standardized β = 0.111, P = 0.001) and femoral neck BMD (standardized β = 0.078, P = 0.021). A first-degree FHD was associated with increased BMD, insulin resistance, and hyperinsulinemia. CONCLUSIONS Our study indicated that normoglycemic postmenopausal women with a first-degree FHD exhibit increased BMD with insulin resistance and hyperinsulinemia. A first-degree FHD was an independent factor associated with elevated BMD in Chinese women after menopause.
Collapse
Affiliation(s)
- Lijuan Yang
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, Zhejiang Province, China
| | | | | | | | | | | |
Collapse
|
32
|
Liu JM, Zhu DL, Mu YM, Xia WB. Management of fracture risk in patients with diabetes-Chinese Expert Consensus. J Diabetes 2019; 11:906-919. [PMID: 31219236 DOI: 10.1111/1753-0407.12962] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/20/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China
| | - Da-Long Zhu
- Department of Endocrinology, Nanjing Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Yi-Ming Mu
- Department of Endocrinology, The General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Wei-Bo Xia
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
33
|
Zhang L, Li P, Tang Z, Dou Q, Feng B. Effects of GLP-1 receptor analogue liraglutide and DPP-4 inhibitor vildagliptin on the bone metabolism in ApoE -/- mice. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:369. [PMID: 31555683 DOI: 10.21037/atm.2019.06.74] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background It has been reported that glucagon-like peptide-1 (GLP-1) can alleviate diabetic osteoporosis (DOP). This study was to investigate the effects of GLP-1RA liraglutide and dipeptidyl peptdase-4 (DPP-4) inhibitor vildagliptin on the advanced glycation end products (AGEs)-induced bone injury in ApoE-/- mice with euglycemia. Methods The bone markers OC, PINP, PTH, TRACP and CTX, the mRNA and protein expressions of RAGE in the femur, and the femoral morphology index were determined to evaluate whether the osteoporosis was improved by liraglutide or vildagliptin. Results AGEs adversely affected the bone metabolism, characterized by reduced OC and increased CTX. However, vildagliptin reduced AGEs and increased OC, and liraglutide significantly decreased AGEs and PTH. Both vildagliptin and liraglutide had no effects on the bone metrology and RAGE expression in the femurs of ApoE-/- mice. Conclusions The elevated AGEs may exacerbate osteogenesis and increase bone resorption, and vildagliptin/liraglutide may improve bone metabolism.
Collapse
Affiliation(s)
- Lizhi Zhang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Peicheng Li
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhaosheng Tang
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qin Dou
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bo Feng
- Department of Endocrinology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
34
|
The Link Between Bone Osteocalcin and Energy Metabolism in a Group of Postmenopausal Women. CURRENT HEALTH SCIENCES JOURNAL 2019; 45:47-51. [PMID: 31297262 PMCID: PMC6592666 DOI: 10.12865/chsj.45.01.06] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/16/2019] [Indexed: 01/10/2023]
Abstract
There is a dual relationship between bone and tissues involved in energy metabolism (fat tissue and beta-pancreatic cells). Thus, bone remodeling is an energy consuming process, but osteocalcin, the main on-collagenic protein, synthesized by osteoblas during bone formation exerts a number of biological effects on beta-pancreatic and adipose cells. With this data, we wanted to see if the presence of a chronic metabolic disorder such as type 2 diabetes mellitus (T2DM) influence this complex dual relationship. For this, we conducted a cross-sectional study to evaluate the relation between osteocalcin and energetic metabolism in a group of 146 postmenopausal womens with and without T2DM at CI Parhon National Institute of Endocrinology, Bucharest. Clinical, metabolic and hormonal parameters were evaluated. For statistical analysis we used Student t-test and the Spearman correlation (statistical significance: p <0.05). Results: 63 patients with T2DM (63.88±8.56 years) and 83 women in the control group (60.21±8.77 years) were included. Diabetic women showed a lower level of serum total osteocalcin (p<0.05) HDL-cholesterol (p=0.02), and 25-hydroxyvitamin D (25(OH)D). The body mass index (BMI), glycemic metabolism parameters and triglyceride levels (p<0.05) were higher in this group. We found correlations between osteocalcin and metabolic elements: negative with BMI (r=-0.329, p<0.05), glycated hemoglobin (HbA1c) (r=-0.398, p<0.05), and serum triglycerides (r=-0.329, p<0.05) respectively positive with HDL-cholesterol (r=0.279, p=0.001) for the entire group of patients. Conclusions: Our study indicated the presence of significant correlations between serum osteocalcin and glycemic and lipid metabolism parameters, independent of the presence of diabetes.
Collapse
|
35
|
Roles for osteocalcin in brain signalling: implications in cognition- and motor-related disorders. Mol Brain 2019; 12:23. [PMID: 30909971 PMCID: PMC6434857 DOI: 10.1186/s13041-019-0444-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/13/2019] [Indexed: 12/16/2022] Open
Abstract
It is now generally accepted that the extra-skeleton functionalities of bone are multifaceted. Its endocrine functions came first to light when it was realized that osteoblasts, the bone forming cells, maintain energy homeostasis by improving glucose metabolism, insulin sensitivity and energy expenditure through osteocalcin, a multipurpose osteokine secreted by osteoblasts. Recently, the emerging knowledge on the functional aspects of this osteokine expanded to properties including adult and maternal regulation of cognitive functions. Therapeutic potential of this osteokine has also been recently reported in experimental Parkinson’s disease models. This review highlights such findings on the functions of osteocalcin in the brain and emphasizes on exploring and analyzing much more in-depth basic and clinical studies.
Collapse
|
36
|
Dumitru N, Carsote M, Cocolos A, Petrova E, Olaru M, Caragheorgheopol A, Dumitrache C, Ghemigian A. Metabolic and bone profile in postmenopausal women with and without type 2 diabetes: a cross-sectional study. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2019; 57:61-67. [PMID: 30447149 DOI: 10.2478/rjim-2018-0036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Current studies support the implication of metabolic changes associated with type 2 diabetes in altering bone metabolism, structure and resistance. OBJECTIVE We conducted a cross-sectional study on postmenopausal women aimed to analyze the differences in metabolic and bone profile in patients with and without type 2 diabetes Methods. We analyzed the metabolic and bone profile in postmenopausal women with and without type 2 diabetes (T2DM). Clinical, metabolic, hormonal parameters, along with lumbar, hip and femoral bone mineral density (BMD) and trabecular bone score (TBS) were evaluated. RESULTS 56 women with T2DM(63.57±8.97 years) and 83 non-T2DM (60.21±8.77 years) were included. T2DM patients presented a higher value of body mass index (BMI) and BMD vs. control group (p = 0.001; p = 0.03-lumbar level, p = 0.07-femoral neck and p = 0.001-total hip). Also, BMI correlated positively with lumbar-BMD and glycated hemoglobin (HbA1c) (r = 0.348, p = 0.01; r = 0.269, p = 0.04), correlation maintained even after age and estimated glomerular filtration rate (eGFR) adjustment (r = 0.383, p = 0.005; r = 0.237, p = 0.08). Diabetic patients recorded lower levels of 25(OH)D(p = 0.05), bone markers (p ≤ 0.05) and TBS(p = 0.07). For the entire patient group we found a negative correlation between HbA1c level and bone markers: r = -0.358, p = 0.0005-osteocalcin, r = -0.40, p = 0.0005-P1NP, r = -0.258, p = 0.005-crosslaps. CONCLUSIONS Our results indicate the presence of altered bone microarchitecture in T2DZ patients according to the TBS score, combined with lower levels of bone markers, with a statistically significant negative correlation between HbA1c level and bone markers.
Collapse
Affiliation(s)
- Nicoleta Dumitru
- "C.I. Parhon" National Institute of Endocrinology, Bucharest,Romania
- "Carol Davila" University of Medicine and Pharmacy, Bucharest,Romania
| | - Mara Carsote
- "C.I. Parhon" National Institute of Endocrinology, Bucharest,Romania
- "Carol Davila" University of Medicine and Pharmacy, Bucharest,Romania
| | - Andra Cocolos
- "C.I. Parhon" National Institute of Endocrinology, Bucharest,Romania
- "Carol Davila" University of Medicine and Pharmacy, Bucharest,Romania
| | - Eugenia Petrova
- "C.I. Parhon" National Institute of Endocrinology, Bucharest,Romania
- "Carol Davila" University of Medicine and Pharmacy, Bucharest,Romania
| | - Maria Olaru
- "C.I. Parhon" National Institute of Endocrinology, Bucharest,Romania
| | | | | | - Adina Ghemigian
- "C.I. Parhon" National Institute of Endocrinology, Bucharest,Romania
- "Carol Davila" University of Medicine and Pharmacy, Bucharest,Romania
| |
Collapse
|
37
|
Affiliation(s)
- Nancy J Brown
- From the Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
38
|
Seppä S, Tenhola S, Voutilainen R. Association of Serum Total Osteocalcin Concentrations With Endogenous Glucocorticoids and Insulin Sensitivity Markers in 12-Year-Old Children: A Cross-Sectional Study. Front Endocrinol (Lausanne) 2019; 10:798. [PMID: 31803143 PMCID: PMC6877498 DOI: 10.3389/fendo.2019.00798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 11/01/2019] [Indexed: 11/24/2022] Open
Abstract
Background: Osteocalcin (OC) is an osteoblast-derived marker of bone turnover that has recently been linked to glucose metabolism, glucocorticoid action, and cardiovascular risk. Objective: We determined whether serum total OC (tOC) is associated with cardiometabolic factors, such as insulin sensitivity (IS) markers and endogenous glucocorticoids in 12-year-old children. In addition, we assessed whether low birth weight or exposure to maternal preeclampsia affect tOC concentrations. Methods: In this cross-sectional study, 192 children (109 girls) were studied at 12 years of age. Seventy of them had been born small (SGA), 78 appropriate for gestational age (AGA), and 44 from preeclamptic pregnancies (PRE) as AGA. Blood pressure was measured, and fasting blood samples were collected for markers of glucose metabolism, osteoblast, adipocyte, and adrenocortical function. IS was estimated by Quantitative Insulin Sensitivity Check Index (QUICKI). Free cortisol index (FCI) was calculated as serum cortisol/corticosteroid binding globulin. Results: The highest tOC concentrations were detected in midpubertal children (Tanner B/G stage 3). The children in the highest tOC quartile (n = 48) had lower body mass index (BMI), waist-to-height ratio, diastolic blood pressure, leptin, cortisol/cortisone ratio and FCI, and higher insulin-like growth factor I (IGF-I), IGF-binding protein-1 (IGFBP-1), IGFBP-3, and alkaline phosphatase (ALP) than those in the lower tOC quartiles (p < 0.02 for all). QUICKI was similar in these subgroups. In logistic regression analysis, pubertal developmental stages 2 and 3, high ALP, IGF-I, and low FCI and BMI (p < 0.02 for all) were associated independently with higher tOC. The means of serum tOC and IS markers were similar in the SGA, AGA, and PRE subgroups. Conclusions: In both sexes, the highest tOC levels were detected in midpubertal children reflecting the fast pubertal growth phase. Higher tOC levels were associated with lower BMI and FCI, whereas no association was found with IS. Birth weight or exposure to preeclampsia had no effect on tOC concentrations.
Collapse
Affiliation(s)
- Satu Seppä
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
- *Correspondence: Satu Seppä
| | - Sirpa Tenhola
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
- Department of Pediatrics, Kymenlaakso Central Hospital, Kotka, Finland
| | - Raimo Voutilainen
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
39
|
Yang YY, Zheng SC, Wang WC, Yang ZW, Shan C, Zhang YW, Qi Y, Chen YH, Gu WQ, Wang WQ, Zhao HY, Liu JM, Sun SY. Osteocalcin Levels in Male Idiopathic Hypogonadotropic Hypogonadism: Relationship With the Testosterone Secretion and Metabolic Profiles. Front Endocrinol (Lausanne) 2019; 10:687. [PMID: 31681165 PMCID: PMC6798147 DOI: 10.3389/fendo.2019.00687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/23/2019] [Indexed: 11/13/2022] Open
Abstract
Idiopathic hypogonadotropic hypogonadism (IHH) patients are characterized by the absence of puberty and varying degrees of deteriorated metabolic conditions. Osteocalcin (OC) could regulate testosterone secretion and energy metabolism, but it remains unknown whether such an effect exists in IHH patients. Our study is aimed to examine the relationship between serum OC levels with testosterone and its responsiveness to gonadotropin stimulation and metabolic profiles in male IHH patients. A total of 99 male patients aged 18-37 years and diagnosed with IHH were enrolled in the current study, and the relationships between OC and testicular volume, baseline total testosterone (TT), free testosterone (FT), and peak TT (Tmax) levels after human chorionic gonadotropin (hCG) stimulation, gonadotropin responsiveness index (GRI), which is calculated by dividing Tmax by testicular volume, as well as metabolic profiles, such as 2-h post-challenge glucose (2hPG) and fat percentage (fat%), were analyzed. The results showed that OC had an independent negative relationship with testicular volume (r = -0.253, P = 0.012) and a positive association with Tmax (r = 0.262, P = 0.014) after adjusting for confounders. In addition, OC was a major determinant of GRI (adjusted R 2 for the model = 0.164, P = 0.012), fat% (adjusted R 2 for the model = 0.100, P = 0.004), and 2hPG (adjusted R 2 for the model = 0.054, P = 0.013) in IHH patients. In conclusion, OC is associated with testosterone secretion upon gonadotropin stimulation, glucose metabolism, and fat mass variations in IHH. This study was registered at clinicaltrials.gov (NCT02310074).
Collapse
Affiliation(s)
- Yu-Ying Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Si-Chang Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Wen-Cui Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Zu-Wei Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Chang Shan
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Yu-Wen Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Rui-jin Hospital North, Shanghai, China
| | - Yan Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Rui-jin Hospital North, Shanghai, China
| | - Yu-Hong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Rui-jin Hospital North, Shanghai, China
| | - Wei-Qiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Wei-Qing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Hong-Yan Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
- *Correspondence: Jian-Min Liu
| | - Shou-Yue Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai, China
- Department of Endocrine and Metabolic Diseases, Shanghai Jiao-Tong University School of Medicine, Rui-jin Hospital North, Shanghai, China
- Shou-Yue Sun
| |
Collapse
|
40
|
Guo XZ, Shan C, Hou YF, Zhu G, Tao B, Sun LH, Zhao HY, Ning G, Li ST, Liu JM. Osteocalcin Ameliorates Motor Dysfunction in a 6-Hydroxydopamine-Induced Parkinson's Disease Rat Model Through AKT/GSK3β Signaling. Front Mol Neurosci 2018; 11:343. [PMID: 30319352 PMCID: PMC6170617 DOI: 10.3389/fnmol.2018.00343] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Osteoblasts derived osteocalcin (OCN) is recently reported to be involved in dopaminergic neuronal development. As dopaminergic neuronal injury in the substantia nigra (SN) is a pathological hallmark of Parkinson’s disease (PD), we investigated whether OCN could exert protective effects on 6-hydroxydopamine (6-OHDA)-induced PD rat model. Our data showed that the OCN level in the cerebrospinal fluid (CSF) in PD rat models was significantly lower than that in controls. Intervention with OCN could improve the behavioral dysfunction in PD rat models and reduce the tyrosine hydroxylase (TH) loss in the nigrostriatal system. In addition, OCN could inhibit the astrocyte and microglia proliferation in the SN of PD rats. In vitro studies showed that OCN significantly ameliorated the neurotoxicity of 6-OHDA through the AKT/GSK3β signaling pathway. In summary, OCN plays a protective role against parkinsonian neurodegeneration in the PD rat model, suggesting a potential therapeutic use of OCN in PD.
Collapse
Affiliation(s)
- Xing-Zhi Guo
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Chang Shan
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Yan-Fang Hou
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Geng Zhu
- Bio-X Institutes, Key laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Bei Tao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Li-Hao Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Hong-Yan Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Sheng-Tian Li
- Bio-X Institutes, Key laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| |
Collapse
|
41
|
Massera D, Biggs ML, Walker MD, Mukamal KJ, Ix JH, Djousse L, Valderrábano RJ, Siscovick DS, Tracy RP, Xue X, Kizer JR. Biochemical Markers of Bone Turnover and Risk of Incident Diabetes in Older Women: The Cardiovascular Health Study. Diabetes Care 2018; 41:1901-1908. [PMID: 30002202 PMCID: PMC6105330 DOI: 10.2337/dc18-0849] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/18/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the relationship of osteocalcin (OC), a marker of bone formation, and C-terminal cross-linked telopeptide of type I collagen (CTX), a marker of bone resorption, with incident diabetes in older women. RESEARCH DESIGN AND METHODS The analysis included 1,455 female participants from the population-based Cardiovascular Health Study (CHS) (mean [SD] age 74.6 [5.0] years). The cross-sectional association of serum total OC and CTX levels with insulin resistance (HOMA-IR) was examined using multiple linear regression. The longitudinal association of both markers with incident diabetes, defined by follow-up glucose measurements, medications, and ICD-9 codes, was examined using multivariable Cox proportional hazards models. RESULTS OC and CTX were strongly correlated (r = 0.80). In cross-sectional analyses, significant or near-significant inverse associations with HOMA-IR were observed for continuous levels of OC (β = -0.12 per SD increment; P = 0.004) and CTX (β = -0.08 per SD; P = 0.051) after full adjustment for demographic, lifestyle, and clinical covariates. During a median follow-up of 11.5 years, 196 cases of incident diabetes occurred. After full adjustment, both biomarkers exhibited inverse associations with incident diabetes (OC: hazard ratio 0.85 per SD [95% CI 0.71-1.02; P = 0.075]; CTX: 0.82 per SD [0.69-0.98; P = 0.031]), associations that were comparable in magnitude and approached or achieved statistical significance. CONCLUSIONS In late postmenopausal women, lower OC and CTX levels were associated with similarly increased risks of insulin resistance at baseline and incident diabetes over long-term follow-up. Further research to delineate the mechanisms linking abnormal bone homeostasis and energy metabolism could uncover new approaches for the prevention of these age-related disorders.
Collapse
Affiliation(s)
- Daniele Massera
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| | | | - Marcella D Walker
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | | | - Joachim H Ix
- University of California San Diego, San Diego, CA
| | | | | | | | | | - Xiaonan Xue
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| | - Jorge R Kizer
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| |
Collapse
|
42
|
Marsot C, Chanson P, Borson-Chazot F. [Bone and metabolism]. ANNALES D'ENDOCRINOLOGIE 2018; 79 Suppl 1:S40-S47. [PMID: 30213304 DOI: 10.1016/s0003-4266(18)31236-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Bone is now considered as a particular endocrine organ. Its endocrine function is not yet fully understood and has been the subject of several conferences at the European Society of Endocrinology Congress 2018. Bone regulates phosphate metabolism by secreting fibroblast growth factor 23; it also regulates glucose metabolism via osteocalcin and energy metabolism, thanks to lipocalin 2, a new hormone acting on the brain. In addition, the incidence of diabetes continues to grow, and its impact on bone has been demonstrated, with an increased risk of fractures regardless the type of diabetes. The mechanism of bone fragility in this disease is not fully known but it involves a decrease in bone turnover and bone demineralization. Recent findings on the role of bone on glucose and mineral metabolism could open therapeutic perspectives, especially for the treatment of diabetes or obesity.
Collapse
Affiliation(s)
- Charlotte Marsot
- Fédération d'endocrinologie, diabétologie et maladies métaboliques, hôpital Louis-Pradel, 59, boulevard Pinel, 69500 Bron, France.
| | - Philippe Chanson
- Service d'endocrinologie, diabétologie, métabolisme et nutrition, hôpital Bicêtre, 78, rue du Général-Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Françoise Borson-Chazot
- Fédération d'endocrinologie, diabétologie et maladies métaboliques, hôpital Louis-Pradel, 59, boulevard Pinel, 69500 Bron, France
| |
Collapse
|
43
|
Liu DM, Mosialou I, Liu JM. Bone: Another potential target to treat, prevent and predict diabetes. Diabetes Obes Metab 2018; 20:1817-1828. [PMID: 29687585 DOI: 10.1111/dom.13330] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 12/30/2022]
Abstract
Type 2 diabetes mellitus is now a worldwide health problem with increasing prevalence. Mounting efforts have been made to treat, prevent and predict this chronic disease. In recent years, increasing evidence from mice and clinical studies suggests that bone-derived molecules modulate glucose metabolism. This review aims to summarize our current understanding of the interplay between bone and glucose metabolism and to highlight potential new means of therapeutic intervention. The first molecule recognized as a link between bone and glucose metabolism is osteocalcin (OCN), which functions in its active form, that is, undercarboxylated OCN (ucOC). ucOC acts in promoting insulin expression and secretion, facilitating insulin sensitivity, and favouring glucose and fatty acid uptake and utilization. A second bone-derived molecule, lipocalin2, functions in suppressing appetite in mice through its action on the hypothalamus. Osteocytes, the most abundant cells in bone matrix, are suggested to act on the browning of white adipose tissue and energy expenditure through secretion of bone morphogenetic protein 7 and sclerostin. The involvement of bone resorption in glucose homeostasis has also been examined. However, there is evidence indicating the implication of the receptor activator of nuclear factor κ-B ligand, neuropeptide Y, and other known and unidentified bone-derived factors that function in glucose homeostasis. We summarize recent advances and the rationale for treating, preventing and predicting diabetes by skeleton intervention.
Collapse
Affiliation(s)
- Dong-Mei Liu
- Department of Rheumatology, ZhongShan Hospital, FuDan University, Shanghai, China
| | - Ioanna Mosialou
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-jin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW The goal of this review is to summarize recent findings on marrow adipose tissue (MAT) function and to discuss the possibility of targeting MAT for therapeutic purposes. RECENT FINDINGS MAT is characterized with high heterogeneity which may suggest both that marrow adipocytes originate from multiple different progenitors and/or their phenotype is determined by skeletal location and environmental cues. Close relationship to osteoblasts and heterogeneity suggests that MAT consists of cells representing spectrum of phenotypes ranging from lipid-filled adipocytes to pre-osteoblasts. We propose a term of adiposteoblast for describing phenotypic spectrum of MAT. Manipulating with MAT activity in diseases where impairment in energy metabolism correlates with bone functional deficit, such as aging and diabetes, may be beneficial for both. Paracrine activities of MAT might be considered for treatment of bone diseases. MAT has unrecognized potential, either beneficial or detrimental, to regulate bone homeostasis in physiological and pathological conditions. More research is required to harness this potential for therapeutic purposes.
Collapse
Affiliation(s)
- Beata Lecka-Czernik
- Department of Orthopaedic Research, Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH, 43614, USA.
| | - Sudipta Baroi
- Department of Orthopaedic Research, Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Lance A Stechschulte
- Department of Orthopaedic Research, Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Amit Sopan Chougule
- Department of Orthopaedic Research, Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| |
Collapse
|
45
|
Frost M, Balkau B, Hatunic M, Konrad T, Mingrone G, Højlund K. The relationship between bone turnover and insulin sensitivity and secretion: Cross-sectional and prospective data from the RISC cohort study. Bone 2018; 108:98-105. [PMID: 29305997 DOI: 10.1016/j.bone.2017.12.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/18/2017] [Accepted: 12/29/2017] [Indexed: 01/27/2023]
Abstract
Bone metabolism appears to influence insulin secretion and sensitivity, and insulin promotes bone formation in animals, but similar evidence in humans is limited. The objectives of this study are to explore if bone turnover markers were associated with insulin secretion and sensitivity and to determine if bone turnover markers predict changes in insulin secretion and sensitivity. The study population encompassed 576 non-diabetic adult men with normal glucose tolerance (NGT; n=503) or impaired glucose regulation (IGR; n=73). Baseline markers of bone resorption (CTX) and formation (P1NP) were determined in the fasting state and after a 2-h hyperinsulinaemic, euglycaemic clamp. An intravenous glucose tolerance test (IVGTT) and a 2-h oral glucose tolerance test (OGTT) were performed at baseline, and the OGTT was repeated after 3years. There were no differences in bone turnover marker levels between NGT and IGR. CTX and P1NP levels decreased by 8.0% (p<0.001) and 1.9% (p<0.01) between baseline and steady-state during the clamp. Fasting plasma glucose was inversely associated with CTX and P1NP both before and after adjustment for recruitment centre, age, BMI, smoking and physical activity. However, baseline bone turnover markers were neither associated with insulin sensitivity (assessed using hyperinsulinaemic euglycaemic clamp and OGTT) nor with insulin secretion capacity (based on IVGTT and OGTT) at baseline or at follow-up. Although inverse associations between fasting glucose and markers of bone turnover were identified, this study cannot support an association between insulin secretion and sensitivity in healthy, non-diabetic men.
Collapse
Affiliation(s)
- Morten Frost
- Endocrine Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Beverley Balkau
- CESP, Faculty of Medicine - University Paris-South, Faculty of Medicine - University Versailles-St Quentin, INSERM U1018, University Paris-Saclay, Villejuif, France
| | - Mensud Hatunic
- Endocrinology Department, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Thomas Konrad
- Clinic of Pediatrics I, Johann Wolfgang Goethe Universität am Main, Frankfurt, Germany
| | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University of the Sacred Heart, Rome, Italy; Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Kurt Højlund
- Endocrine Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Section of Molecular Diabetes & Metabolism, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
46
|
Al-Khalidi B, Rotondi MA, Kimball SM, Ardern CI. Clinical utility of serum 25-hydroxyvitamin D in the diagnosis of insulin resistance and estimation of optimal 25-hydroxyvitamin D in U.S. adults. Diabetes Res Clin Pract 2017; 134:80-90. [PMID: 28951344 DOI: 10.1016/j.diabres.2017.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/23/2017] [Accepted: 09/18/2017] [Indexed: 12/25/2022]
Abstract
AIMS To assess the clinical utility of measuring serum 25-hydroxyvitamin-D [25(OH)D] along with traditional risk factors in the diagnosis of insulin resistance (IR) and to estimate the optimal 25(OH)D level associated with normal glucose and insulin homeostasis. METHODS A cross-sectional analysis of 6868 adults aged≥20years without diagnosed diabetes in the National Health and Nutrition Examination Survey, with available standardized 25(OH)D data (2001-2010). IR was defined by the homeostatic-model-assessment of insulin resistance (HOMA-IR; ≥75th percentile, sex-specific: 3.9 in men or 3.6 in women). Using logistic regression, two risk models were developed to estimate the risk of IR: Model 1 included established risk factors, and Model 2 additionally included serum 25(OH)D. Predictiveness curves and decision-curve analysis were used to assess differences in IR detection among models. Receiver-operating-characteristic curves were used to estimate the lower threshold for 25(OH)D. Results were validated in a testing sample. RESULTS Model 2 marginally improved detection of IR: at a risk threshold of 0.2, adding 25(OH)D would identify an additional 2 to 4 cases per 1000 people. Overall, the lower 25(OH)D threshold was estimated at 60nmol/L, however, the threshold differed by ethnicity (Mexican-Americans: 54nmo/L, non-Hispanic whites: 68nmol/L, and non-Hispanic blacks: 41nmol/L). CONCLUSION Addition of serum 25(OH)D to traditional risk factors provided small incremental improvement in detection of IR in asymptomatic adults. The optimal 25(OH)D threshold was estimated to be at least 60nmol/L, however, the threshold may differ by ethnic-background. Further research is needed to validate these results in other populations.
Collapse
Affiliation(s)
- Banaz Al-Khalidi
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.
| | - Michael A Rotondi
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | | | - Chris I Ardern
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Ortinau LC, Linden MA, Dirkes R, Rector RS, Hinton PS. Obesity and type 2 diabetes, not a diet high in fat, sucrose, and cholesterol, negatively impacts bone outcomes in the hyperphagic Otsuka Long Evans Tokushima Fatty rat. Bone 2017; 105:200-211. [PMID: 28893629 DOI: 10.1016/j.bone.2017.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/14/2017] [Accepted: 09/08/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Obesity and type 2 diabetes (T2D) increase fracture risk; however, the association between obesity/T2D may be confounded by consumption of a diet high in fat, sucrose, and cholesterol (HFSC). OBJECTIVE The study objective was to determine the main and interactive effects of obesity/T2D and a HFSC diet on bone outcomes using hyperphagic Otuska Long Evans Tokushima Fatty (OLETF) rats and normophagic Long Evans Tokushima Otsuka (LETO) controls. METHODS At 8weeks of age, male OLETF and LETO rats were randomized to either a control (CON, 10 en% from fat as soybean oil) or HFSC (45 en% from fat as soybean oil/lard, 17 en% sucrose, and 1wt%) diet, resulting in four treatment groups. At 32weeks, total body bone mineral content (BMC) and density (BMD) and body composition were measured by dual-energy X-ray absorptiometry, followed by euthanasia and collection of blood and tibiae. Bone turnover markers and sclerostin were measured using ELISA. Trabecular microarchitecture of the proximal tibia and geometry of the tibia mid-diaphysis were measured using microcomputed tomography; whole-bone and tissue-level biomechanical properties were evaluated using torsional loading of the tibia. Two-factor ANOVA was used to determine main and interactive effects of diet (CON vs. HFSC) and obesity/T2D (OLETF vs. LETO) on bone outcomes. RESULTS Hyperphagic OLEFT rats had greater final body mass, body fat, and fasting glucose than normophagic LETO, with no effect of diet. Total body BMC and serum markers of bone formation were decreased, and bone resorption and sclerostin were increased in obese/T2D OLETF rats. Trabecular bone volume and microarchitecture were adversely affected by obesity/T2D, but not diet. Whole-bone and tissue-level biomechanical properties of the tibia were not affected by obesity/T2D; the HFSC diet improved biomechanical properties only in LETO rats. CONCLUSIONS Obesity/T2D, regardless of diet, negatively impacted the balance between bone formation and resorption and trabecular bone volume and microarchitecture in OLETF rats.
Collapse
Affiliation(s)
- Laura C Ortinau
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Melissa A Linden
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States; Research Service-Harry S Truman Memorial Veterans Medical Center, Columbia, MO, United States
| | - Rebecca Dirkes
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States; Department of Medicine, Gastroenterology and Hepatology, University of Missouri, Columbia, MO, United States; Research Service-Harry S Truman Memorial Veterans Medical Center, Columbia, MO, United States
| | - Pamela S Hinton
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
48
|
Colleluori G, Napoli N, Phadnis U, Armamento-Villareal R, Villareal DT. Effect of Weight Loss, Exercise, or Both on Undercarboxylated Osteocalcin and Insulin Secretion in Frail, Obese Older Adults. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4807046. [PMID: 28951766 PMCID: PMC5603129 DOI: 10.1155/2017/4807046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/25/2017] [Accepted: 07/06/2017] [Indexed: 11/17/2022]
Abstract
BACKGROUND Obesity exacerbates age-related decline in glucometabolic control. Undercarboxylated osteocalcin (UcOC) regulates pancreatic insulin secretion. The long-term effect of lifestyle interventions on UcOC and insulin secretion has not been investigated. METHODS One hundred seven frail, obese older adults were randomized into the control (N = 27), diet (N = 26), exercise (N = 26), and diet-exercise (N = 28) groups for 1 year. Main outcomes included changes in UcOC and disposition index (DI). RESULTS UcOC increased in the diet group (36 ± 11.6%) but not in the other groups (P < 0.05 between groups). Although similar increases in DI occurred in the diet-exercise and diet groups at 6 months, DI increased more in the diet-exercise group (92.4 ± 11.4%) than in the diet group (61.9 ± 15.3%) at 12 months (P < 0.05). UcOC and body composition changes predicted DI variation in the diet group only (R2 = 0.712), while adipocytokines and physical function changes contributed to DI variation in both the diet (∆R2 = 0.140 and 0.107) and diet-exercise (∆R2 = 0.427 and 0.243) groups (P < 0.05 for all). CONCLUSIONS Diet, but not exercise or both, increases UcOC, whereas both diet and diet-exercise increase DI. UcOC accounts for DI variation only during active weight loss, while adipocytokines and physical function contribute to diet-exercise-induced DI variation, highlighting different mechanisms for lifestyle-induced improvements in insulin secretion. This trial was registered with ClinicalTrials.gov number NCT00146107.
Collapse
Affiliation(s)
- Georgia Colleluori
- Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- University Campus-Biomedico, 00128, Rome, Italy
| | | | - Uma Phadnis
- Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Reina Armamento-Villareal
- Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Dennis T. Villareal
- Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| |
Collapse
|
49
|
The association between the baseline bone resorption marker CTX and incident dysglycemia after 4 years. Bone Res 2017; 5:17020. [PMID: 28698818 PMCID: PMC5496471 DOI: 10.1038/boneres.2017.20] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 01/01/2017] [Accepted: 02/27/2017] [Indexed: 12/29/2022] Open
Abstract
Bone is an endocrine organ involved in modulating glucose homeostasis. The role of the bone formation marker osteocalcin (OCN) in predicting diabetes was reported, but with conflicting results. No study has explored the association between baseline bone resorption activity and incident diabetes or prediabetes during follow-up. Our objective was to examine the relationship between the baseline bone resorption marker crosslinked C-telopeptide of type I collagen (CTX) and glycemic dysregulation after 4 years. This longitudinal study was conducted in a university teaching hospital. A total of 195 normal glucose tolerant (NGT) women at baseline were invited for follow-up. The incidence of diabetes and prediabetes (collectively defined as dysglycemia) was recorded. A total of 128 individuals completed the 4-year study. The overall conversion rate from NGT to dysglycemia was 31.3%. The incidence of dysglycemia was lowest in the middle tertile [16.3% (95% confidence interval (CI), 6.8%–30.7%)] compared with the lower [31.0% (95% CI, 17.2%–46.1%)] and upper [46.5% (95% CI, 31.2%–62.6%)] tertiles of CTX, with a significant difference seen between the middle and upper tertiles (P=0.002 5). After adjusting for multiple confounding variables, the upper tertile of baseline CTX was associated with an increased risk of incident dysglycemia, with an odds ratio of 7.09 (95% CI, 1.73–28.99) when the middle tertile was the reference. Osteoclasts actively regulate glucose homeostasis in a biphasic model that moderately enhanced bone resorption marker CTX at baseline provides protective effects against the deterioration of glucose metabolism, whereas an overactive osteoclastic function contributes to an increased risk of subsequent dysglycemia.
Collapse
|