1
|
Smith C, Lin X, Parker L, Yeap BB, Hayes A, Levinger I. The role of bone in energy metabolism: A focus on osteocalcin. Bone 2024; 188:117238. [PMID: 39153587 DOI: 10.1016/j.bone.2024.117238] [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: 04/08/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Understanding the mechanisms involved in whole body glucose regulation is key for the discovery of new treatments for type 2 diabetes (T2D). Historically, glucose regulation was largely focused on responses to insulin and glucagon. Impacts of incretin-based therapies, and importance of muscle mass, are also highly relevant. Recently, bone was recognized as an endocrine organ, with several bone proteins, known as osteokines, implicated in glucose metabolism through their effects on the liver, skeletal muscle, and adipose tissue. Research efforts mostly focused on osteocalcin (OC) as a leading example. This review will provide an overview on this role of bone by discussing bone turnover markers (BTMs), the receptor activator of nuclear factor kB ligand (RANKL), osteoprotegerin (OPG), sclerostin (SCL) and lipocalin 2 (LCN2), with a focus on OC. Since 2007, some, but not all, research using mostly OC genetically modified animal models suggested undercarboxylated (uc) OC acts as a hormone involved in energy metabolism. Most data generated from in vivo, ex vivo and in vitro models, indicate that exogenous ucOC administration improves whole-body and skeletal muscle glucose metabolism. Although data in humans are generally supportive, findings are often discordant likely due to methodological differences and observational nature of that research. Overall, evidence supports the concept that bone-derived factors are involved in energy metabolism, some having beneficial effects (ucOC, OPG) others negative (RANKL, SCL), with the role of some (LCN2, other BTMs) remaining unclear. Whether the effect of osteokines on glucose regulation is clinically significant and of therapeutic value for people with insulin resistance and T2D remains to be confirmed.
Collapse
Affiliation(s)
- Cassandra Smith
- Nutrition & Health Innovation Research Institute, School of Health and Medical Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Perth, Western Australia, Australia; Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St Albans, VIC, Australia
| | - Xuzhu Lin
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Lewan Parker
- Institute for Physical Activity and Nutrition (IPAN), Deakin University, Geelong, VIC, Australia
| | - Bu B Yeap
- Medical School, The University of Western Australia, Perth, Western Australia, Australia; Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia
| | - Alan Hayes
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St Albans, VIC, Australia; Department of Medicine - Western Health, The University of Melbourne, Footscray, VIC, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Victoria University and Western Health, St Albans, VIC, Australia; Department of Medicine - Western Health, The University of Melbourne, Footscray, VIC, Australia.
| |
Collapse
|
2
|
Liu YF, Tian Y, Chen XF, Zhang C, Huang L. Role of osteokines in atherosclerosis. Cell Biochem Funct 2024; 42:e4107. [PMID: 39154288 DOI: 10.1002/cbf.4107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/03/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
Despite their diverse physiologies and roles, the heart, skeletal muscles, and smooth muscles all derive from a common embryonic source as bones. Moreover, bone tissue, skeletal and smooth muscles, and the heart share conserved signaling pathways. The maintenance of skeletal health is precisely regulated by osteocytes, osteoblasts, and osteoclasts through coordinated secretion of bone-derived factors known as osteokines. Increasing evidence suggests the involvement of osteokines in regulating atherosclerotic vascular disease. Therefore, this review aims to examine the evidence for the role of osteokines in atherosclerosis development and progression comprehensively. Specifically discussed are extensively studied osteokines in atherosclerosis such as osteocalcin, osteopontin, osteoprotegerin, and fibroblast growth factor 23. Additionally, we highlighted the effects of exercise on modulating these key regulators derived from bone tissue metabolism. We believe that gaining an enhanced understanding of how osteocalcin contributes to the process of atherosclerosis will enable us to develop targeted and comprehensive therapeutic strategies against diseases associated with its progression.
Collapse
Affiliation(s)
- Yi-Fan Liu
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, People's Republic of China
| | - Xiao-Fang Chen
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, People's Republic of China
| | - Liang Huang
- Institute of Translational Medicine, School of Basic Medical, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
3
|
Deng AF, Wang FX, Wang SC, Zhang YZ, Bai L, Su JC. Bone-organ axes: bidirectional crosstalk. Mil Med Res 2024; 11:37. [PMID: 38867330 PMCID: PMC11167910 DOI: 10.1186/s40779-024-00540-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
In addition to its recognized role in providing structural support, bone plays a crucial role in maintaining the functionality and balance of various organs by secreting specific cytokines (also known as osteokines). This reciprocal influence extends to these organs modulating bone homeostasis and development, although this aspect has yet to be systematically reviewed. This review aims to elucidate this bidirectional crosstalk, with a particular focus on the role of osteokines. Additionally, it presents a unique compilation of evidence highlighting the critical function of extracellular vesicles (EVs) within bone-organ axes for the first time. Moreover, it explores the implications of this crosstalk for designing and implementing bone-on-chips and assembloids, underscoring the importance of comprehending these interactions for advancing physiologically relevant in vitro models. Consequently, this review establishes a robust theoretical foundation for preventing, diagnosing, and treating diseases related to the bone-organ axis from the perspective of cytokines, EVs, hormones, and metabolites.
Collapse
Affiliation(s)
- An-Fu Deng
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fu-Xiao Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Si-Cheng Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200444, China
| | - Ying-Ze Zhang
- Department of Orthopaedics, the Third Hospital of Hebei Medical University, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, 050051, China.
| | - Long Bai
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, Zhejiang, China.
| | - Jia-Can Su
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
4
|
Sinder SB, Sharma SV, Shirvaikar IS, Pradhyumnan H, Patel SH, Cabeda Diaz I, Perez GG, Bramlett HM, Raval AP. Impact of menopause-associated frailty on traumatic brain injury. Neurochem Int 2024; 176:105741. [PMID: 38621511 DOI: 10.1016/j.neuint.2024.105741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Navigating menopause involves traversing a complex terrain of hormonal changes that extend far beyond reproductive consequences. Menopausal transition is characterized by a decrease in estradiol-17β (E2), and the impact of menopause resonates not only in the reproductive system but also through the central nervous system, musculoskeletal, and gastrointestinal domains. As women undergo menopausal transition, they become more susceptible to frailty, amplifying the risk and severity of injuries, including traumatic brain injury (TBI). Menopause triggers a cascade of changes leading to a decline in muscle mass, accompanied by diminished tone and excitability, thereby restricting the availability of irisin, a crucial hormone derived from muscles. Concurrently, bone mass undergoes reduction, culminating in the onset of osteoporosis and altering the dynamics of osteocalcin, a hormone originating from bones. The diminishing levels of E2 during menopause extend their influence on the gut microbiota, resulting in a reduction in the availability of tyrosine, tryptophan, and serotonin metabolites, affecting neurotransmitter synthesis and function. Understanding the interplay between menopause, frailty, E2 decline, and the intricate metabolisms of bone, gut, and muscle is imperative when unraveling the nuances of TBI after menopause. The current review underscores the significance of accounting for menopause-associated frailty in the incidence and consequences of TBI. The review also explores potential mechanisms to enhance gut, bone, and muscle health in menopausal women, aiming to mitigate frailty and improve TBI outcomes.
Collapse
Affiliation(s)
- Sophie B Sinder
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sabrina V Sharma
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Isha S Shirvaikar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Hari Pradhyumnan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Shahil H Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Indy Cabeda Diaz
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Gina G Perez
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| |
Collapse
|
5
|
Saadon SR, Allwsh TA. Lipocalin-2 as a marker of inflammation, bone density, and triglyceride-glucose index for new-onset arthritis patients in Mosul, Iraq. Qatar Med J 2024; 2024:23. [PMID: 38765271 PMCID: PMC11097680 DOI: 10.5339/qmj.2024.23] [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: 11/08/2023] [Accepted: 04/17/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVE Lipocalin-2 is an acute phase-associated adipokine that can serve as an inflammatory and biomarker indicator of cartilage deterioration in osteoarthritis. However, its role in the musculoskeletal system remains not fully understood. Hence, this study aimed to evaluate lipocalin-2 and its relationship with markers of inflammation (Interferon-gamma, ESR, and CRP), bone density (vitamin D3 and calcium), and the triglyceride-glucose index in new-onset arthritis patients in Mosul, Iraq. METHODS This study included 125 participants aged 20 to 65, divided into two groups. The Arthritis Patient Group comprised 70 participants (37 females and 33 males) attending the Bone Diseases Consultation Unit at the Ibn Sina Teaching Hospital in Mosul, Iraq. The Control Group comprised 31 females and 24 males. Ethical approval was obtained from the Iraqi Ministry of Health - Nineveh Health (No. 2022095). Commercial ELISA kits were used to measure serum lipocalin-2, Interferon-gamma, ESR, and CRP as inflammation markers, vitamin D3, and calcium as bone density markers. Moreover, the Triglyceride Glucose (TYG) Index was evaluated. RESULTS The findings revealed a significant increase in lipocalin-2 levels in males compared to females, with LCN-2 increasing with age. Arthritis patients showed a significant increase (72%) in lipocalin-2 levels. Inflammatory indicators (erythrocyte sedimentation rate, C-reactive protein, interferon-gamma) displayed significant increases (46%, 1200%, and 581%, respectively). Glucose (23%), triglycerides (71%), and TYG index (21%) also exhibited significant increases. Meanwhile, bone density indicators (vitamin D3 and calcium) found a significant decrease (53% and 20%, respectively) in arthritis patients. Linear correlation coefficient (R) analysis revealed a significant positive relationship between lipocalin-2 and indicators of inflammation, glucose, TG, and TYG index. CONCLUSION This study's findings suggest that LCN-2 serum levels were higher in patients with new-onset arthritis than in controls in Mosul, and LCN-2 serum increased in males compared with females and getting older serum LCN-2 increased for the patients and control groups. Furthermore, a significant correlation was found between the Triglyceride Glucose Index, which measures metabolic disorders, and serum LCN-2 levels and inflammatory indicators in new-onset arthritis patients in Mosul, Iraq.
Collapse
Affiliation(s)
- Safa Rabea Saadon
- Department of Chemistry, Collage of Science, University of Mosul, Mosul, Iraq *
| | - Thikra Ali Allwsh
- Department of Chemistry, Collage of Science, University of Mosul, Mosul, Iraq *
| |
Collapse
|
6
|
Zhao Y, Peng X, Wang Q, Zhang Z, Wang L, Xu Y, Yang H, Bai J, Geng D. Crosstalk Between the Neuroendocrine System and Bone Homeostasis. Endocr Rev 2024; 45:95-124. [PMID: 37459436 DOI: 10.1210/endrev/bnad025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Indexed: 01/05/2024]
Abstract
The homeostasis of bone microenvironment is the foundation of bone health and comprises 2 concerted events: bone formation by osteoblasts and bone resorption by osteoclasts. In the early 21st century, leptin, an adipocytes-derived hormone, was found to affect bone homeostasis through hypothalamic relay and the sympathetic nervous system, involving neurotransmitters like serotonin and norepinephrine. This discovery has provided a new perspective regarding the synergistic effects of endocrine and nervous systems on skeletal homeostasis. Since then, more studies have been conducted, gradually uncovering the complex neuroendocrine regulation underlying bone homeostasis. Intriguingly, bone is also considered as an endocrine organ that can produce regulatory factors that in turn exert effects on neuroendocrine activities. After decades of exploration into bone regulation mechanisms, separate bioactive factors have been extensively investigated, whereas few studies have systematically shown a global view of bone homeostasis regulation. Therefore, we summarized the previously studied regulatory patterns from the nervous system and endocrine system to bone. This review will provide readers with a panoramic view of the intimate relationship between the neuroendocrine system and bone, compensating for the current understanding of the regulation patterns of bone homeostasis, and probably developing new therapeutic strategies for its related disorders.
Collapse
Affiliation(s)
- Yuhu Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaole Peng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Qing Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Zhiyu Zhang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Liangliang Wang
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
- Department of Orthopedics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230022, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University; Orthopedics Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
7
|
Lu N, Shan C, Fu JR, Zhang Y, Wang YY, Zhu YC, Yu J, Cai J, Li SX, Tao T, Liu W. RANKL Is Independently Associated with Increased Risks of Non-Alcoholic Fatty Liver Disease in Chinese Women with PCOS: A Cross-Sectional Study. J Clin Med 2023; 12:jcm12020451. [PMID: 36675380 PMCID: PMC9864426 DOI: 10.3390/jcm12020451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Women with polycystic ovarian syndrome (PCOS) are more likely to have non-alcoholic fatty liver disease (NAFLD) than non-PCOS women; however, the exact mechanism underlying this trend is unknown. The receptor activator of NF-κB ligand (RANKL) is strongly involved in bone metabolism and has multiple functions. Recent studies suggest that RANKL is implicated in hepatic insulin resistance (IR), which is the highest risk factor for NAFLD. This study aimed to assess the role of RANKL in NAFLD in Chinese women with PCOS. A cross-sectional observational study was conducted on women newly diagnosed with PCOS, which included 146 patients with NAFLD and 142 patients without NAFLD. Sex hormones, glucose, insulin, and lipids were measured, and anthropometric data were collected. The concentration of serum total RANKL was measured using commercial ELISA kits. PCOS patients with NAFLD had a significantly higher mean age, body mass index (BMI), waist circumference (WC), and worsened metabolic profile than non-NAFLD subjects. The concentrations of high-sensitivity C-reactive protein, total cholesterol, and low-density lipoprotein cholesterol increased with the RANKL tertile (p for trend = 0.023, 0.026, and 0.035, respectively). A significantly positive association was found between RANKL (per SD change) and the risks of NAFLD (OR = 1.545, 95% CI = 1.086−2.199) after adjusting for confounders, including demographic factors, metabolic markers, and sex hormones. Subgroup multivariate logistic analyses stratified by age, BMI, and WC showed the same tendency. In addition, the positive association between RANKL and NAFLD seemed more prominent in lean patients with a BMI < 24 kg/m2 (OR = 1.70, 95% CI = 1.06−2.75) when compared to overweight/obesity subjects. Therefore, this study suggests that RANKL is positively associated with the increased risk of NAFLD in Chinese women with PCOS, independent of metabolic and reproductive factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tao Tao
- Correspondence: (T.T.); (W.L.)
| | - Wei Liu
- Correspondence: (T.T.); (W.L.)
| |
Collapse
|
8
|
Hue I, Capilla E, Rosell-Moll E, Balbuena-Pecino S, Goffette V, Gabillard JC, Navarro I. Recent advances in the crosstalk between adipose, muscle and bone tissues in fish. Front Endocrinol (Lausanne) 2023; 14:1155202. [PMID: 36998471 PMCID: PMC10043431 DOI: 10.3389/fendo.2023.1155202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
Control of tissue metabolism and growth involves interactions between organs, tissues, and cell types, mediated by cytokines or direct communication through cellular exchanges. Indeed, over the past decades, many peptides produced by adipose tissue, skeletal muscle and bone named adipokines, myokines and osteokines respectively, have been identified in mammals playing key roles in organ/tissue development and function. Some of them are released into the circulation acting as classical hormones, but they can also act locally showing autocrine/paracrine effects. In recent years, some of these cytokines have been identified in fish models of biomedical or agronomic interest. In this review, we will present their state of the art focusing on local actions and inter-tissue effects. Adipokines reported in fish adipocytes include adiponectin and leptin among others. We will focus on their structure characteristics, gene expression, receptors, and effects, in the adipose tissue itself, mainly regulating cell differentiation and metabolism, but in muscle and bone as target tissues too. Moreover, lipid metabolites, named lipokines, can also act as signaling molecules regulating metabolic homeostasis. Regarding myokines, the best documented in fish are myostatin and the insulin-like growth factors. This review summarizes their characteristics at a molecular level, and describes both, autocrine effects and interactions with adipose tissue and bone. Nonetheless, our understanding of the functions and mechanisms of action of many of these cytokines is still largely incomplete in fish, especially concerning osteokines (i.e., osteocalcin), whose potential cross talking roles remain to be elucidated. Furthermore, by using selective breeding or genetic tools, the formation of a specific tissue can be altered, highlighting the consequences on other tissues, and allowing the identification of communication signals. The specific effects of identified cytokines validated through in vitro models or in vivo trials will be described. Moreover, future scientific fronts (i.e., exosomes) and tools (i.e., co-cultures, organoids) for a better understanding of inter-organ crosstalk in fish will also be presented. As a final consideration, further identification of molecules involved in inter-tissue communication will open new avenues of knowledge in the control of fish homeostasis, as well as possible strategies to be applied in aquaculture or biomedicine.
Collapse
Affiliation(s)
- Isabelle Hue
- Laboratory of Fish Physiology and Genomics, UR1037, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Rennes, France
| | - Encarnación Capilla
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Enrique Rosell-Moll
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Sara Balbuena-Pecino
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Valentine Goffette
- Laboratory of Fish Physiology and Genomics, UR1037, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Rennes, France
| | - Jean-Charles Gabillard
- Laboratory of Fish Physiology and Genomics, UR1037, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Rennes, France
| | - Isabel Navarro
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Burgos-Gamez X, Morales-Castillo P, Fernandez-Mejia C. Maternal adaptations of the pancreas and glucose homeostasis in lactation and after lactation. Mol Cell Endocrinol 2023; 559:111778. [PMID: 36162635 DOI: 10.1016/j.mce.2022.111778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/04/2022] [Accepted: 09/13/2022] [Indexed: 02/03/2023]
Abstract
During lactation, the maternal physiology adapts to bear the nutritional requirements of the offspring. The exocrine and endocrine pancreas are central to nutrient handling, promoting digestion and metabolism. In concert with prolactin, insulin is a determinant factor for milk synthesis. The investigation of the pancreas during lactation has been scattered over several periods. The investigations that laid the foundation of lactating pancreatic physiology and glucose homeostasis were conducted in the decades of 1970-1980. With the development of molecular biology, newer studies have revealed the molecular mechanisms involved in the endocrine pancreas during breastfeeding. There has been a surge of information recently about unexpected changes in the pancreas at the end of the lactation period and after weaning. In this review, we aim to gather information on the changes in the pancreas and glucose homeostasis during and after lactation and discuss the outcomes derived from the current discoveries.
Collapse
Affiliation(s)
- Xadeni Burgos-Gamez
- Unidad de Genética de la Nutrición. Instituto de Investigaciones Biomédicas. Universidad Nacional Autónoma de México/ Instituto Nacional de Pediatría. Avenida del Iman#1, 4th floor, Mexico City, 04500, Mexico
| | - Paulina Morales-Castillo
- Unidad de Genética de la Nutrición. Instituto de Investigaciones Biomédicas. Universidad Nacional Autónoma de México/ Instituto Nacional de Pediatría. Avenida del Iman#1, 4th floor, Mexico City, 04500, Mexico
| | - Cristina Fernandez-Mejia
- Unidad de Genética de la Nutrición. Instituto de Investigaciones Biomédicas. Universidad Nacional Autónoma de México/ Instituto Nacional de Pediatría. Avenida del Iman#1, 4th floor, Mexico City, 04500, Mexico.
| |
Collapse
|
10
|
Tarantino U, Greggi C, Visconti VV, Cariati I, Bonanni R, Gasperini B, Nardone I, Gasbarra E, Iundusi R. Sarcopenia and bone health: new acquisitions for a firm liaison. Ther Adv Musculoskelet Dis 2022; 14:1759720X221138354. [PMID: 36465879 PMCID: PMC9716454 DOI: 10.1177/1759720x221138354] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/23/2022] [Indexed: 11/15/2023] Open
Abstract
Osteosarcopenia (OS) is a newly defined condition represented by the simultaneous presence of osteopenia/osteoporosis and sarcopenia, the main age-related diseases. The simultaneous coexistence of the two phenotypes derives from the close connection of the main target tissues involved in their pathogenesis: bone and muscle. These two actors constitute the bone-muscle unit, which communicates through a biochemical and mechanical crosstalk which involves multiple factors. Altered pattern of molecular pathways leads to an impairment of both the functionality of the tissue itself and the communication with the complementary tissue, composing the OS pathogenesis. Recent advances in the genetics field have provided the opportunity to delve deeper into the complex biological and molecular mechanisms underlying OS. Unfortunately, there are still many gaps in our understanding of these pathways, but it has proven essential to apply strategies such as exercise and nutritional intervention to counteract OS. New therapeutic strategies that simultaneously target bone and muscle tissue are limited, but recently new targets for the development of dual-action drug therapies have been identified. This narrative review aims to provide an overview of the latest scientific evidence associated with OS, a complex disorder that will pave the way for future research aimed at understanding the bone-muscle-associated pathogenetic mechanisms.
Collapse
Affiliation(s)
- Umberto Tarantino
- Department of Clinical Sciences and
Translational Medicine, University of Rome ‘Tor Vergata’, Rome, Italy
- Department of Orthopedics and Traumatology, PTV
Foundation, Rome, Italy
| | - Chiara Greggi
- Department of Clinical Sciences and
Translational Medicine, University of Rome ‘Tor Vergata’, Rome, Italy
| | - Virginia Veronica Visconti
- Department of Clinical Sciences and
Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1,
00133 Rome, Italy
| | - Ida Cariati
- Department of Biomedicine and Prevention,
University of Rome ‘Tor Vergata’, Rome, Italy
| | - Roberto Bonanni
- Department of Biomedicine and Prevention,
University of Rome ‘Tor Vergata’, Rome, Italy
| | - Beatrice Gasperini
- Department of Biomedicine and Prevention,
University of Rome ‘Tor Vergata’, Rome, Italy
| | - Italo Nardone
- Department of Orthopedics and Traumatology, PTV
Foundation, Rome, Italy
| | - Elena Gasbarra
- Department of Orthopedics and Traumatology, PTV
Foundation, Rome, Italy
| | - Riccardo Iundusi
- Department of Orthopedics and Traumatology,
PTV Foundation, Rome, Italy
| |
Collapse
|
11
|
Kumar V, Bolanthakodi N, Vidyasagar S, Holla A, Sheik SM, Abhishek S. Association of serum osteocalcin with beta cell function, insulin resistance, and glycemic parameters in south Indian type 2 diabetic subjects. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01087-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
12
|
Osteocalcin Alleviates Lipopolysaccharide-Induced Acute Inflammation via Activation of GPR37 in Macrophages. Biomedicines 2022; 10:biomedicines10051006. [PMID: 35625743 PMCID: PMC9138386 DOI: 10.3390/biomedicines10051006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
The G protein-coupled receptor 37 (GPR37) has been reported to be expressed in macrophages and the activation of GPR37 by its ligand/agonist, and it can regulate macrophage-associated functions and inflammatory responses. Since our previous work identified that osteocalcin (OCN) acts as an endogenous ligand for GPR37 and can elicit various intracellular signals by interacting with GPR37, we thus hypothesized that OCN may also play a functional role in macrophage through the activation of GPR37. To verify the hypothesis, we conducted a series of in vivo and in vitro studies in lipopolysaccharide (LPS)-challenged mice and primary cultured macrophages. Our results reveal that the OCN gene deletion (OCN−/−) and wild type (WT) mice showed comparable death rates and inflammatory cytokines productions in response to a lethal dose of LPS exposure. However, the detrimental effects caused by LPS were significantly ameliorated by exogenous OCN treatments in both WT and OCN−/− mice. Notably, the protective effects of OCN were absent in GPR37−/− mice. In coordination with the in vivo results, our in vitro studies further illustrated that OCN triggered intracellular responses via GPR37 in peritoneal macrophages by regulating the release of inflammatory factors and macrophage phagocytic function. Finally, we exhibited that the adoptive transfer of OCN-treated macrophages from WT mice significantly inhibits the release of pro-inflammatory cytokines in GPR37−/− mice exposed to LPS. Taken together, these findings suggest a protective role of OCN against LPS-caused acute inflammation, by the activation of GPR37 in macrophages, and provide a potential application of the activation of the OCN/GPR37 regulatory axis as a therapeutic strategy for inflammatory diseases.
Collapse
|
13
|
Goit RK, Taylor AW, Yin Lo AC. The central melanocortin system as a treatment target for obesity and diabetes: A brief overview. Eur J Pharmacol 2022; 924:174956. [DOI: 10.1016/j.ejphar.2022.174956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022]
|
14
|
Song L, Li H, Liu Y, Zhang X, Wen Y, Zhang K, Zhang M. Postnatal deletion of β-catenin in preosteoblasts regulates global energy metabolism through increasing bone resorption and adipose tissue fibrosis. Bone 2022; 156:116320. [PMID: 34973494 DOI: 10.1016/j.bone.2021.116320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 11/02/2022]
Abstract
Many studies revealed bone can regulate global energy metabolism and our previous study also showed that Wnt/β-catenin pathway is involved in this process. To better understand the participation of canonical Wnt pathway in energy metabolism, we examined the β-catenin knock-out (β-cat KO) mice by crossing the osterix-cre transgenic mice with β-cateninflox/flox mice. First, we identified that postnatal deletion of β-catenin in preosteoblasts led to decreased fat mass and increased energy expenditure in mice. Osteoprotegerin administration largely rescued the decreased fat mass and partly normalized the energy expenditure accompanied by the inhibition of bone resorption. Anti-resorption with alendronate or RANKL-antibody could also partly rescued the decreased bone mass, decreased fat mass and increased energy expenditure in β-cat KO mice. We further found that the adipose cells in the inguinal fat tissue were smaller and the extracellular matrix components around adipocytes accumulated more in β-cat KO mice than their controls by histomorphology. Gene analysis by RT-PCR showed that the expression of collagen VI is 4.8 folds in adipose tissue of the β-cat KO mice compared with the control mice. We further detected the expression of cytokines which were related to fibrosis and the data showed that the level of TGF-beta1 was elevated in both of bone marrow serum and adipose tissue derived from the β-cat KO mice. After administration of TGF-beta1 neutralizing antibody, the impaired energy metabolism was partly rescued in β-cat KO mice. Besides, anti-resorption treatment and TGF-beta1 antibody could partly suppress the increased expression of genes related to fat tissue fibrosis. These results indicate that the abnormal global energy metabolism in β-cat KO mice may be attributed to increasing bone resorption and adipose tissue fibrosis.
Collapse
Affiliation(s)
- Lige Song
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Huijuan Li
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yichen Liu
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiaoya Zhang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yuhua Wen
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Keqin Zhang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Mingzhu Zhang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 1 Dongjiao Minxiang, West Area, Beijing, 100730, China.
| |
Collapse
|
15
|
Travis C, Srivastava PS, Hawke TJ, Kalaitzoglou E. Diabetic Bone Disease and Diabetic Myopathy: Manifestations of the Impaired Muscle-Bone Unit in Type 1 Diabetes. J Diabetes Res 2022; 2022:2650342. [PMID: 35601019 PMCID: PMC9119786 DOI: 10.1155/2022/2650342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/18/2022] [Accepted: 04/28/2022] [Indexed: 11/25/2022] Open
Abstract
Type 1 diabetes is associated with complications affecting muscle and bone, with diabetic bone disease and diabetic myopathy becoming increasingly reported in the past few decades. This review is aimed at succinctly reviewing the literature on the current knowledge regarding these increasingly identified and possibly interconnected complications on the musculoskeletal system. Furthermore, this review summarizes several nonmechanical factors that could be mediating the development and progression of premature musculoskeletal decline in this population and discusses preventative measures to reduce the burden of diabetes on the musculoskeletal system.
Collapse
Affiliation(s)
- Callie Travis
- University of Kentucky College of Medicine, Lexington, KY, USA
| | - Priya S. Srivastava
- Department of Pediatrics, Division of Pediatric Endocrinology, UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Thomas J. Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Evangelia Kalaitzoglou
- University of Kentucky, Barnstable Brown Diabetes Center, Lexington, KY, USA
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| |
Collapse
|
16
|
Xu C, Gong M, Wen S, Zhou M, Li Y, Zhou L. The Comparative Study on the Status of Bone Metabolism and Thyroid Function in Diabetic Patients with or without Ketosis or Ketoacidosis. Diabetes Metab Syndr Obes 2022; 15:779-797. [PMID: 35309734 PMCID: PMC8926020 DOI: 10.2147/dmso.s349769] [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: 11/23/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE This study aims to identify changes in bone turnover markers and thyroid function in diabetic ketosis (DK) and diabetic ketoacidosis (DKA). MATERIALS AND METHODS We compared data from the Department of Endocrinology at Shanghai Pudong Hospital from 2018 to 2020 on the pancreatic status and previous glucose control, bone transformation, calcium homeostasis, and thyroid function in groups with diabetes (DM alone, n=602), DK (n=232), and DKA (n=60). Similar comparisons were made in recurrent DK (A) (n=17) and single DK (A) (n=272). RESULTS The fasting C-peptide level decreased significantly, but hemoglobin A1c (HbA1c) levels were higher in DK or DKA (p<0.05). Blood calcium and 25-hydroxyvitamin D3 (25-OH-VitD3) levels were significantly lower in DKA (p<0.05), but parathyroid hormone (PTH) levels remained constant across all three groups. The N-terminal middle molecular fragment of osteocalcin (N-MID) and β-C terminal cross-linking telopeptide of type 1 collagen (β-CTX) showed significant inverse alterations in DKA, regardless of gender or age (p<0.05). Otherwise, DKA significantly inhibited thyroid function (p<0.05). Furthermore, Spearman correlation analyses revealed a relationship between N-MID and HbA1c in DM alone (r=-0.27, p<0.01), while total triiodothyronine (TT3, r=0.62, p<0.01) or free T3 (FT3, r=0.61, p<0.01) in DK, and DKA (TT3, r=0.45, p<0.01; FT3, r=0.43, p<0.01). Multilinear regression analyses revealed that β-CTX (β=0.564), HbA1c (β=-0.196), TT3 (β=0.183), and 25-OH-VitD3 (β=-0.120) were the only independent determinants of N-MID in DM, whereas FT3 (β=0.491), β-CTX (β=0.315) in DK, and FT3 (β=0.420), β-CTX (β=0.367), TG (β=-0.278) in DKA. Only 25-OH-VitD3 was found to be significantly lower in recurrent DK (A) than in single onset DK (A) (p<0.05), and β-CTX (β=0.745) was found to be significantly independently associated with N-MID. CONCLUSION Our preliminary findings show a dramatic change in bone turnover markers in DM patients with DK and DKA, and this change may be related to thyroid function.
Collapse
Affiliation(s)
- Chenglin Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, People’s Republic of China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, People’s Republic of China
| | - Mingyue Zhou
- Clinical Research OB/GYN REI Division, University of California, San Francisco, CA, USA
| | - Yanyan Li
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Shanghai, People’s Republic of China
- Correspondence: Ligang Zhou, Tel +8613611927616, Email
| |
Collapse
|
17
|
Qian Z, Li H, Yang H, Yang Q, Lu Z, Wang L, Chen Y, Li X. Osteocalcin attenuates oligodendrocyte differentiation and myelination via GPR37 signaling in the mouse brain. SCIENCE ADVANCES 2021; 7:eabi5811. [PMID: 34678058 PMCID: PMC8535816 DOI: 10.1126/sciadv.abi5811] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/01/2021] [Indexed: 05/24/2023]
Abstract
The bone-derived hormone osteocalcin (OCN) is crucial for brain development and neural cognitive functions, yet the exact roles of OCN in central nervous system (CNS) remain elusive. Here, we find that genetic deletion of OCN facilitates oligodendrocyte (OL) differentiation and hypermyelination in the CNS. Although dispensable for the proliferation of oligodendrocyte precursor cells (OPCs), OCN is critical for the myelination of OLs, which affects myelin production and remyelination after demyelinating injury. Genome-wide RNA sequencing analyses reveal that OCN regulates a number of G protein–coupled receptors and myelination-associated transcription factors, of which Myrf might be a key downstream effector in OLs. GPR37 is identified as a previously unknown receptor for OCN, thus regulating OL differentiation and CNS myelination. Overall, these findings suggest that OCN orchestrates the transition between OPCs and myelinating OLs via GPR37 signaling, and hence, the OCN/GPR37 pathway regulates myelin homeostasis in the CNS.
Collapse
Affiliation(s)
- Zhengjiang Qian
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hongchao Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Haiyang Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qin Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhonghua Lu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liping Wang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, Fujian, China
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Center for Excellence in Brain Science and Intelligence Technology, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
18
|
Song L, Huang Y, Long J, Li Y, Pan Z, Fang F, Long Y, Huang C, Qi N, Guo Y, Xia H, Jiang Y. The Role of Osteocalcin in Placental Function in Gestational Diabetes Mellitus. Reprod Biol 2021; 21:100566. [PMID: 34626941 DOI: 10.1016/j.repbio.2021.100566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 10/20/2022]
Abstract
Evidence for the role of osteocalcin in glucose metabolism is increasing. The aim of this study was to examine the associations between osteocalcin and gestational diabetes mellitus. Thirteen discovery study subjects and 76 reduplication study subjects were recruited from the Maternal and Child Health Hospital Guangxi Zhuang Autonomous Region from May 2018 to August 2018. Total osteocalcin and biochemical indices of maternal serum and umbilical vein serum were analyzed. Placental tissue samples were used for transcriptome sequencing. For the discovery study subjects, the total osteocalcin concentration in umbilical vein serum was significantly higher than that in maternal serum and umbilical artery serum (55.32 ng/mL ± 17.37 vs. 12.06 ng/mL ± 5.42 [P < 0.001] vs. 38.31 ng/mL ± 11.52 [P < 0.01]), suggesting that trophoblasts may synthesize osteocalcin. In a reduplication subject study, the gestational diabetes mellitus group had lower umbilical vein serum total osteocalcin (51.46 ng/mL ± 24.29 vs. 67.00 ng/mL ± 25.33, P = 0.008), lower adiponectin (1099.72 μg/L ± 102.65 vs. 1235.85 μg/L ± 94.63, P < 0.001). Spearman's correlation analysis showed that umbilical vein serum total osteocalcin levels were closely correlated with leptin (r = -0.456, P = 0.007). A coexpression model of the placental RNA sequence was constructed. Two modules were correlated with osteocalcin, and the Gene ontology pathways of these modules were rich in glucose and lipid metabolism. In conclusion, the placenta may synthesize osteocalcin by itself, and a lower osteocalcin level in umbilical vein serum is associated with gestational diabetes mellitus.
Collapse
Affiliation(s)
- Liang Song
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yiming Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Junqing Long
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yuanfan Li
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Zongqin Pan
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Fang Fang
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yu Long
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Cheng Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Nana Qi
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yajie Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Hongwei Xia
- Department of Obstetrics, The Maternity Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China.
| | - Yonghua Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; Guangxi Key Laboratory of Genomic and Personalized Medicine, Nanning, Guangxi, 530021, China; Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi, 530021, China.
| |
Collapse
|
19
|
Zhang W, Liu P, Ling S, Wang F, Wang S, Chen T, Zhou R, Xia X, Yao Z, Fan Y, Wang N, Wang J, Tucker HO, Guo X. Forkhead box P1 (Foxp1) in osteoblasts regulates bone mass accrual and adipose tissue energy metabolism. J Bone Miner Res 2021; 36:2017-2026. [PMID: 34131944 DOI: 10.1002/jbmr.4394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/07/2021] [Accepted: 06/12/2021] [Indexed: 11/08/2022]
Abstract
Adiponectin (AdipoQ), a hormone abundantly secreted by adipose tissues, has multiple beneficial functions, including insulin sensitization as well as lipid and glucose metabolism. It has been reported that bone controls energy metabolism through an endocrine-based mechanism. In this study, we observed that bone also acts as an important endocrine source for AdipoQ, and its capacity in osteoblasts is controlled by the forkhead box P1 (FOXP1) transcriptional factor. Deletion of the Foxp1 gene in osteoblasts led to augmentation of AdipoQ levels accompanied by fueled energy expenditure in adipose tissues. In contrast, overexpression of Foxp1 in bones impaired AdipoQ secretion and restrained energy consumption. Chromatin immunoprecipitation sequencing (ChIP-seq) analysis revealed that AdipoQ expression, which increases as a function of bone age, is directly controlled by FOXP1. Our results indicate that bones, especially aged bones, provide an important source of a set of endocrine factors, including AdipoQ, that control body metabolism. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Wei Zhang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Pei Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Shifeng Ling
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Fuhua Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Shaojiao Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Tienan Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Rujiang Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Xuechun Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Zhengju Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Ying Fan
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiqiu Wang
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haley O Tucker
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - Xizhi Guo
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Takaya J, Tanabe Y, Kaneko K. Increased lipocalin 2 levels in adolescents with type 2 diabetes mellitus. J Pediatr Endocrinol Metab 2021; 34:979-985. [PMID: 34118796 DOI: 10.1515/jpem-2021-0216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/02/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Bone can act as an endocrine organ through the secretion of bone-specific hormones, i.e., osteokines. Recent research has demonstrated that lipocalin 2 (LCN2) secreted by osteoblasts are part of an important endocrine system that is finely tuned with other organs to ensure homeostatic balance and health. The aim of this study was to explore the association between bone and glucose metabolism in adolescents with obesity and type 2 diabetes mellitus (DM2). METHODS The participants were 8 adolescents with DM2 (5 males, 3 females; age: 17.0 (14.0-20.0) years, median (interquartile range)), 14 adolescents with simple obesity (9 males, 5 females; age: 13.5 (12.4-15.5) years), and 15 controls (6 males, 9 females; age: 13.3 (11.0-15.0) years). Serum LCN2 and under-carboxylated osteocalcin (un-OC) levels were measured using enzyme-linked immunosorbent assays. RESULTS The LCN2 levels were higher in patients with DM2 (58.1 (34.2-95.0) ng/mL; median (interquartile range)), but not in those with obesity (30.8 (23.1-38.3) ng/mL), when compared to the controls (18.2 (9.8-25.7) ng/mL). In the whole study group overall, serum LCN2 was positively correlated with the Model Assessment of Insulin Resistance score (r=0.339, p=0.046) and body mass index (r=0.580, p<0.0001), and negatively correlated with adiponectin (r=-0.462, p=0.005). A multiple stepwise regression model showed that serum adiponectin was an independent predictor of serum LCN2. CONCLUSIONS The results of this study indicate that further investigations are warranted to determine whether LCN2 may act as a sensitive indicator of early-stage insulin resistance.
Collapse
Affiliation(s)
- Junji Takaya
- Department of Pediatrics, Kawachi General Hospital, Higashi-Osaka, Osaka, 578-0954, Japan.,Department of Pediatrics, Kansai Medical University, Hirakata, Osaka, Japan
| | - Yuko Tanabe
- Department of Pediatrics, Kansai Medical University, Hirakata, Osaka, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
21
|
Cariati I, Bonanni R, Onorato F, Mastrogregori A, Rossi D, Iundusi R, Gasbarra E, Tancredi V, Tarantino U. Role of Physical Activity in Bone-Muscle Crosstalk: Biological Aspects and Clinical Implications. J Funct Morphol Kinesiol 2021; 6:55. [PMID: 34205747 PMCID: PMC8293201 DOI: 10.3390/jfmk6020055] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Bone and muscle tissues influence each other through the integration of mechanical and biochemical signals, giving rise to bone-muscle crosstalk. They are also known to secrete osteokines, myokines, and cytokines into the circulation, influencing the biological and pathological activities in local and distant organs and cells. In this regard, even osteoporosis and sarcopenia, which were initially thought to be two independent diseases, have recently been defined under the term "osteosarcopenia", to indicate a synergistic condition of low bone mass with muscle atrophy and hypofunction. Undoubtedly, osteosarcopenia is a major public health concern, being associated with high rates of morbidity and mortality. The best current defence against osteosarcopenia is prevention based on a healthy lifestyle and regular exercise. The most appropriate type, intensity, duration, and frequency of exercise to positively influence osteosarcopenia are not yet known. However, combined programmes of progressive resistance exercises, weight-bearing impact exercises, and challenging balance/mobility activities currently appear to be the most effective in optimising musculoskeletal health and function. Based on this evidence, the aim of our review was to summarize the current knowledge about the role of exercise in bone-muscle crosstalk, highlighting how it may represent an effective alternative strategy to prevent and/or counteract the onset of osteosarcopenia.
Collapse
Affiliation(s)
- Ida Cariati
- PhD in Medical-Surgical Biotechnologies and Translational Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy;
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bonanni
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (V.T.)
| | - Federica Onorato
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (F.O.); (A.M.); (D.R.); (R.I.); (E.G.)
| | - Ambra Mastrogregori
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (F.O.); (A.M.); (D.R.); (R.I.); (E.G.)
| | - Danilo Rossi
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (F.O.); (A.M.); (D.R.); (R.I.); (E.G.)
| | - Riccardo Iundusi
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (F.O.); (A.M.); (D.R.); (R.I.); (E.G.)
| | - Elena Gasbarra
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (F.O.); (A.M.); (D.R.); (R.I.); (E.G.)
| | - Virginia Tancredi
- Department of Systems Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (V.T.)
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Umberto Tarantino
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, Viale Oxford 81, 00133 Rome, Italy; (F.O.); (A.M.); (D.R.); (R.I.); (E.G.)
| |
Collapse
|
22
|
Tauer JT, Boraschi-Diaz I, Al Rifai O, Rauch F, Ferron M, Komarova SV. Male but not female mice with severe osteogenesis imperfecta are partially protected from high-fat diet-induced obesity. Mol Genet Metab 2021; 133:211-221. [PMID: 33814269 DOI: 10.1016/j.ymgme.2021.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/16/2021] [Accepted: 03/24/2021] [Indexed: 01/07/2023]
Abstract
Previously we have shown that young mice with a dominant severe form of osteogenesis imperfecta (OI), caused by mutated collagen type I, exhibit an altered glucose/insulin metabolism and energy expenditure along with elevated levels of osteocalcin, a bone-derived hormone involved in the regulation of whole-body metabolism. This study aimed to examine the long-term effects of a western diet in these OI mice. Male and female OI mice and wild type littermates (WT) were fed a high-fat diet (HFD) or a matched low-fat diet (LFD) for 26 weeks. HFD-induced obesity was observed in male and female WT and female OI mice, but not in male OI mice. HFD-fed WT and OI mice of both sexes developed hyperglycemia and glucose intolerance, but the degree of glucose intolerance was significantly lower in male and female OI mice compared to sex- and diet-matched WT mice. Indirect calorimetry revealed increased movement of male OI mice on HFD compared to LFD and, while HFD lowered energy expenditure in WT mice, energy expenditure was not changed in OI mice. Further, HFD-fed male OI mice demonstrated a diet-induced increased expression of the thermogenesis genes, Ucp1 and Pgc1α, in brown adipose tissue. On LFD, total and Gla-13 osteocalcin levels were similar in 30-week-old WT and OI mice, but on HFD, both were significantly higher in OI mice than WT. Thus, male OI mice respond to HFD with increased movement, energy expenditure, brown adipose tissue thermogenesis, and higher levels of osteocalcin, resulting in partial protection against HFD-induced obesity.
Collapse
Affiliation(s)
- Josephine T Tauer
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children-Canada, Montreal, Quebec, Canada.
| | - Iris Boraschi-Diaz
- Shriners Hospital for Children-Canada, Montreal, Quebec, Canada; Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Omar Al Rifai
- Unité de Recherche en Physiologie Moléculaire, Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
| | - Frank Rauch
- Shriners Hospital for Children-Canada, Montreal, Quebec, Canada; Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Mathieu Ferron
- Unité de Recherche en Physiologie Moléculaire, Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada; Départements de Médecine et de Biochimie et Médecine Moléculaire, Université de Montréal, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| |
Collapse
|
23
|
The Role of Vitamin K in Humans: Implication in Aging and Age-Associated Diseases. Antioxidants (Basel) 2021; 10:antiox10040566. [PMID: 33917442 PMCID: PMC8067486 DOI: 10.3390/antiox10040566] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/02/2021] [Accepted: 04/02/2021] [Indexed: 12/17/2022] Open
Abstract
As human life expectancy is rising, the incidence of age-associated diseases will also increase. Scientific evidence has revealed that healthy diets, including good fats, vitamins, minerals, or polyphenolics, could have antioxidant and anti-inflammatory activities, with antiaging effects. Recent studies demonstrated that vitamin K is a vital cofactor in activating several proteins, which act against age-related syndromes. Thus, vitamin K can carboxylate osteocalcin (a protein capable of transporting and fixing calcium in bone), activate matrix Gla protein (an inhibitor of vascular calcification and cardiovascular events) and carboxylate Gas6 protein (involved in brain physiology and a cognitive decline and neurodegenerative disease inhibitor). By improving insulin sensitivity, vitamin K lowers diabetes risk. It also exerts antiproliferative, proapoptotic, autophagic effects and has been associated with a reduced risk of cancer. Recent research shows that protein S, another vitamin K-dependent protein, can prevent the cytokine storm observed in COVID-19 cases. The reduced activation of protein S due to the pneumonia-induced vitamin K depletion was correlated with higher thrombogenicity and possibly fatal outcomes in COVID-19 patients. Our review aimed to present the latest scientific evidence about vitamin K and its role in preventing age-associated diseases and/or improving the effectiveness of medical treatments in mature adults ˃50 years old.
Collapse
|
24
|
Mosialou I, Shikhel S, Luo N, Petropoulou PI, Panitsas K, Bisikirska B, Rothman NJ, Tenta R, Cariou B, Wargny M, Sornay-Rendu E, Nickolas T, Rubin M, Confavreux CB, Kousteni S. Lipocalin-2 counteracts metabolic dysregulation in obesity and diabetes. J Exp Med 2021; 217:151926. [PMID: 32639539 PMCID: PMC7537391 DOI: 10.1084/jem.20191261] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 03/28/2020] [Accepted: 05/15/2020] [Indexed: 12/30/2022] Open
Abstract
Regulation of food intake is a recently identified endocrine function of bone that is mediated by Lipocalin-2 (LCN2). Osteoblast-secreted LCN2 suppresses appetite and decreases fat mass while improving glucose metabolism. We now show that serum LCN2 levels correlate with insulin levels and β-cell function, indices of healthy glucose metabolism, in obese mice and obese, prediabetic women. However, LCN2 serum levels also correlate with body mass index and insulin resistance in the same individuals and are increased in obese mice. To dissect this apparent discrepancy, we modulated LCN2 levels in mice. Silencing Lcn2 expression worsens metabolic dysfunction in genetic and diet-induced obese mice. Conversely, increasing circulating LCN2 levels improves metabolic parameters and promotes β-cell function in mouse models of β-cell failure acting as a growth factor necessary for β-cell adaptation to higher metabolic load. These results indicate that LCN2 up-regulation is a protective mechanism to counteract obesity-induced glucose intolerance by decreasing food intake and promoting adaptive β-cell proliferation.
Collapse
Affiliation(s)
- Ioanna Mosialou
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Steven Shikhel
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Na Luo
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | | | - Konstantinos Panitsas
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Brygida Bisikirska
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Nyanza J Rothman
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Roxane Tenta
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Bertrand Cariou
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, l'Institut du thorax, Nantes, France
| | - Matthieu Wargny
- Université de Nantes, Centre Hospitalier Universitaire Nantes, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, l'Institut du thorax, Nantes, France
| | - Elisabeth Sornay-Rendu
- Institut national de la santé et de la recherche médicale Unités Mixtes de Recherche 1033, Université de Lyon, Hospices Civils de Lyon, Lyon, France
| | - Thomas Nickolas
- Department of Medicine Nephrology, Columbia University Medical Center, New York, NY
| | - Mishaela Rubin
- Department of Medicine Endocrinology, Columbia University Medical Center, New York, NY
| | - Cyrille B Confavreux
- Institut national de la santé et de la recherche médicale Unités Mixtes de Recherche 1033, Université de Lyon, Hospices Civils de Lyon, Lyon, France
| | - Stavroula Kousteni
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| |
Collapse
|
25
|
Al Rifai O, Julien C, Lacombe J, Faubert D, Lira-Navarrete E, Narimatsu Y, Clausen H, Ferron M. The half-life of the bone-derived hormone osteocalcin is regulated through O-glycosylation in mice, but not in humans. eLife 2020; 9:61174. [PMID: 33284103 PMCID: PMC7822592 DOI: 10.7554/elife.61174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022] Open
Abstract
Osteocalcin (OCN) is an osteoblast-derived hormone with pleiotropic physiological functions. Like many peptide hormones, OCN is subjected to post-translational modifications (PTMs) which control its activity. Here, we uncover O-glycosylation as a novel PTM present on mouse OCN and occurring on a single serine (S8) independently of its carboxylation and endoproteolysis, two other PTMs regulating this hormone. We also show that O-glycosylation increases OCN half-life in plasma ex vivo and in the circulation in vivo. Remarkably, in human OCN (hOCN), the residue corresponding to S8 is a tyrosine (Y12), which is not O-glycosylated. Yet, the Y12S mutation is sufficient to O-glycosylate hOCN and to increase its half-life in plasma compared to wildtype hOCN. These findings reveal an important species difference in OCN regulation, which may explain why serum concentrations of OCN are higher in mouse than in human. Bones provide support and protection for organs in the body. However, over the last 15 years researchers have discovered that bones also release chemicals known as hormones, which can travel to other parts of the body and cause an effect. The cells responsible for making bone, known as osteoblasts, produce a hormone called osteocalcin which communicates with a number of different organs, including the pancreas and brain. When osteocalcin reaches the pancreas, it promotes the release of another hormone called insulin which helps regulate the levels of sugar in the blood. Osteocalcin also travels to other organs such as muscle, where it helps to degrade fats and sugars that can be converted into energy. It also has beneficial effects on the brain, and has been shown to aid memory and reduce depression. Osteocalcin has largely been studied in mice where levels are five to ten times higher than in humans. But it is unclear why this difference exists or how it alters the role of osteocalcin in humans. To answer this question, Al Rifai et al. used a range of experimental techniques to compare the structure and activity of osteocalcin in mice and humans. The experiments showed that mouse osteocalcin has a group of sugars attached to its protein structure, which prevent the hormone from being degraded by an enzyme in the blood. Human osteocalcin has a slightly different protein sequence and is therefore unable to bind to this sugar group. As a result, the osteocalcin molecules in humans are less stable and cannot last as long in the blood. Al Rifai et al. showed that when human osteocalcin was modified so the sugar group could attach, the hormone was able to stick around for much longer and reach higher levels when added to blood in the laboratory. These findings show how osteocalcin differs between human and mice. Understanding this difference is important as the effects of osteocalcin mean this hormone can be used to treat diabetes and brain disorders. Furthermore, the results reveal how the stability of osteocalcin could be improved in humans, which could potentially enhance its therapeutic effect.
Collapse
Affiliation(s)
- Omar Al Rifai
- Molecular Physiology Research unit, Institut de Recherches Cliniques de Montréal, Montréal, Canada.,Programme de biologie moléculaire, Université de Montréal, Montréal, Canada
| | - Catherine Julien
- Molecular Physiology Research unit, Institut de Recherches Cliniques de Montréal, Montréal, Canada
| | - Julie Lacombe
- Molecular Physiology Research unit, Institut de Recherches Cliniques de Montréal, Montréal, Canada
| | - Denis Faubert
- Proteomics Discovery Platform, Institut de Recherches Cliniques de Montréal, Montréal, Canada
| | - Erandi Lira-Navarrete
- University of Copenhagen, Faculty of Health Sciences, Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Copenhagen, Denmark
| | - Yoshiki Narimatsu
- University of Copenhagen, Faculty of Health Sciences, Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Copenhagen, Denmark
| | - Henrik Clausen
- University of Copenhagen, Faculty of Health Sciences, Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Copenhagen, Denmark
| | - Mathieu Ferron
- Molecular Physiology Research unit, Institut de Recherches Cliniques de Montréal, Montréal, Canada.,Programme de biologie moléculaire, Université de Montréal, Montréal, Canada.,Département de Médecine, Université de Montréal, Montréal, Canada.,Division of Experimental Medicine, McGill University, Montréal, Canada
| |
Collapse
|
26
|
Yang C, Pan H, Shen L. Pan-Cancer Analyses Reveal Prognostic Value of Osteomimicry Across 20 Solid Cancer Types. Front Mol Biosci 2020; 7:576269. [PMID: 33240930 PMCID: PMC7678014 DOI: 10.3389/fmolb.2020.576269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/12/2020] [Indexed: 11/16/2022] Open
Abstract
Background Osteomimicry of cancer cells had been widely reported in prostate cancer and breast cancer. However, the prognostic value of osteomimicry in various cancer types remained unclear. We hypothesized that osteomimicry would result in remodeling of the tumor microenvironment and was eligible to predict patient prognosis. Methods A comprehensive transcriptomic analysis of the osteomimicry, which was characterized by mRNA expression of SPARC, SPP1, and BGLAP, across 20 solid tumors (7564 patients) using RNA-seq data from The Cancer Genome Atlas (TCGA) was conducted. Samples of each cancer type were classified into subgroups (high vs. low) based on median value of osteomimetic markers, the associations of these markers with clinical outcomes, immune cell infiltration and immune checkpoints expression were explored. Results Each osteomimetic marker harbored prognostic value in the pan-cancer analyses [SPARC: hazard ratio (HR) = 1.10, p = 0.028; SPP1: HR = 1.25, p < 0.001; BGLAP: HR = 1.13, p = 0.005]. Patients with high expression of all the three genes also had significantly unfavorable survival (HR = 1.61, p < 0.0001) compared with those of low expression. Correlation analyses demonstrated that osteomimicry was closely related to tumor purity, dendritic cells (DC) infiltration and expression of immune checkpoints. Conclusion Osteomimicry had prognostic value in various cancer types and the underlying mechanism might correlate to the trapping and dysfunction of DCs in the tumor microenvironment, revealing the potential of osteomimicry as a target of immunotherapy.
Collapse
Affiliation(s)
- Changsheng Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Orthopaedic Hospital of Guangdong Province, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| | - Hehai Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lujun Shen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
Hendriks M, Ramasamy SK. Blood Vessels and Vascular Niches in Bone Development and Physiological Remodeling. Front Cell Dev Biol 2020; 8:602278. [PMID: 33330496 PMCID: PMC7729063 DOI: 10.3389/fcell.2020.602278] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
Recent advances in our understanding of blood vessels and vascular niches in bone convey their critical importance in regulating bone development and physiology. The contribution of blood vessels in bone functions and remodeling has recently gained enormous interest because of their therapeutic potential. The mammalian skeletal system performs multiple functions in the body to regulate growth, homeostasis and metabolism. Blood vessels provide support to various cell types in bone and maintain functional niches in the bone marrow microenvironment. Heterogeneity within blood vessels and niches indicate the importance of specialized vascular niches in regulating skeletal functions. In this review, we discuss physiology of bone vasculature and their specialized niches for hematopoietic stem cells and mesenchymal progenitor cells. We provide clinical and experimental information available on blood vessels during physiological bone remodeling.
Collapse
Affiliation(s)
- Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, London, United Kingdom
| | - Saravana K. Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
28
|
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: 1.0] [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
|
29
|
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: 6] [Impact Index Per Article: 1.5] [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
|
30
|
Kristiansson A, Gram M, Flygare J, Hansson SR, Åkerström B, Storry JR. The Role of α 1-Microglobulin (A1M) in Erythropoiesis and Erythrocyte Homeostasis-Therapeutic Opportunities in Hemolytic Conditions. Int J Mol Sci 2020; 21:ijms21197234. [PMID: 33008134 PMCID: PMC7582998 DOI: 10.3390/ijms21197234] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023] Open
Abstract
α1-microglobulin (A1M) is a small protein present in vertebrates including humans. It has several physiologically relevant properties, including binding of heme and radicals as well as enzymatic reduction, that are used in the protection of cells and tissue. Research has revealed that A1M can ameliorate heme and ROS-induced injuries in cell cultures, organs, explants and animal models. Recently, it was shown that A1M could reduce hemolysis in vitro, observed with several different types of insults and sources of RBCs. In addition, in a recently published study, it was observed that mice lacking A1M (A1M-KO) developed a macrocytic anemia phenotype. Altogether, this suggests that A1M may have a role in RBC development, stability and turnover. This opens up the possibility of utilizing A1M for therapeutic purposes in pathological conditions involving erythropoietic and hemolytic abnormalities. Here, we provide an overview of A1M and its potential therapeutic effect in the context of the following erythropoietic and hemolytic conditions: Diamond-Blackfan anemia (DBA), 5q-minus myelodysplastic syndrome (5q-MDS), blood transfusions (including storage), intraventricular hemorrhage (IVH), preeclampsia (PE) and atherosclerosis.
Collapse
Affiliation(s)
- Amanda Kristiansson
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden;
- Correspondence:
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, 221 84 Lund, Sweden;
| | - Johan Flygare
- Department of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden;
| | - Stefan R. Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences Lund, Lund University, 221 84 Lund, Sweden;
| | - Bo Åkerström
- Section for Infection Medicine, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Jill R. Storry
- Division of Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden;
- Department of Clinical Immunology and Transfusion Medicine, Office of Medical Services, 221 85 Lund, Sweden
| |
Collapse
|
31
|
Yang Y, Zhong W, Huang J, Geng L, Feng Q. Association of serum osteocalcin levels with glucose metabolism in trauma patients. Medicine (Baltimore) 2020; 99:e21901. [PMID: 32899020 PMCID: PMC7478502 DOI: 10.1097/md.0000000000021901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Osteocalcin (OC) is an endocrine hormone that regulates glucose metabolism.The aim of this study was to investigate the relationship between serum OC levels and glucose metabolism after trauma.This was a retrospective study of trauma patients admitted to the Department of Emergency Medicine between October 2017 and April 2019. Age, height, weight, injury severity score, and previous medical history were recorded. Serum N-terminal mid-fragment of OC (N-MID OC), hemoglobin Alc (HbA1c), fasting plasma glucose (FPG), fasting insulin (FINS), C-peptide, and other biochemical indicators were measured. Differences between the HbA1c-L (HbA1c <6.5%) and HbA1c-H (HbA1c ≥6.5%) groups were compared. The association of N-MID OC with indicators of glucose metabolism was analyzed.Out of 394 trauma patients, leukocyte and FPG levels in the HbA1c-H group (n = 93) were higher (P < .05), while N-MID OC levels were lower (P = .011) than the HbA1c-L group (n = 301). N-MID OC was negatively correlated with HbA1c in the total population (r = -0.273, P < .001) as well as in the HbA1c-L (r = -0.289, P < .001) and HbA1c-H (r = -0.390, P < 0.001) groups, and was positively correlated with C-peptide in the HbA1c-H group (r = 0.395, P < .001). The different quartiles in the HbA1c-L showed that N-MID OC declined with increasing HbA1c, which was higher than N-MID OC levels in the HbA1c-H group. Multiple linear regression analysis revealed that serum HbA1c was independently associated with serum OC levels after trauma (β=-1.608, P < .001).This study strongly suggests the importance of serum OC on glucose metabolism in trauma patients. HbA1c is independently associated with serum OC levels.
Collapse
|
32
|
Turcotte AF, Grenier-Larouche T, Lacombe J, Carreau AM, Carpentier AC, Mac-Way F, Tchernof A, Richard D, Biertho L, Lebel S, Marceau S, Ferron M, Gagnon C. Association between changes in bioactive osteocalcin and glucose homeostasis after biliopancreatic diversion. Endocrine 2020; 69:526-535. [PMID: 32419080 DOI: 10.1007/s12020-020-02340-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/04/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE Bone may regulate glucose homeostasis via uncarboxylated bioactive osteocalcin (ucOCN). This study explored whether changes in ucOCN and bone remodeling are associated with change in glucose homeostasis after biliopancreatic diversion (BPD). METHODS In this secondary exploratory analysis of a 1-year prospective observational study, 16 participants (11 men/5 women; 69% with type 2 diabetes; mean BMI 49.4 kg/m2) were assessed before, 3 days, 3 months and 12 months after BPD. Changes in plasma ucOCN and bone markers (C-terminal telopeptide (CTX), total osteocalcin (OCN)) were correlated with changes in insulin resistance or sensitivity indices (HOMA-IR; adipose tissue insulin resistance index (ADIPO-IR) and insulin sensitivity index (SI) from the hyperinsulinemic-euglycemic clamp), insulin secretion rate (ISR) from the hyperglycemic clamp, and disposition index (DI: SI × ISR) using Spearman correlations before and after adjustment for weight loss. RESULTS ucOCN was unchanged at 3 days but increased dramatically at 3 months (+257%) and 12 months (+498%). Change in ucOCN correlated significantly with change in CTX at 3 months (r = 0.62, p = 0.015) and 12 months (r = 0.64, p = 0.025) before adjustment for weight loss. It also correlated significantly with change in fasting insulin (r = -0.53, p = 0.035), HOMA-IR (r = -0.54, p = 0.033) and SI (r = 0.52, p = 0.041) at 3 days, and ADIPO-IR (r = -0.69, p = 0.003) and HbA1c (r = -0.69, p = 0.005) at 3 months. Change in OCN did not correlate with any glucose homeostasis indices. Results were similar after adjustment for weight loss. CONCLUSION The increase in ucOCN may be associated with the improvement in insulin resistance after BPD, independently of weight loss. These findings need to be confirmed in larger, less heterogeneous populations.
Collapse
Affiliation(s)
- Anne-Frédérique Turcotte
- Endocrinology and Nephrology Unit, CHU de Québec-Université Laval Research Centre, Québec City, QC, Canada
- Department of Medicine, Laval University, Québec City, QC, Canada
| | - Thomas Grenier-Larouche
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Julie Lacombe
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada
| | - Anne-Marie Carreau
- Endocrinology and Nephrology Unit, CHU de Québec-Université Laval Research Centre, Québec City, QC, Canada
- Department of Medicine, Laval University, Québec City, QC, Canada
| | - André C Carpentier
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Fabrice Mac-Way
- Endocrinology and Nephrology Unit, CHU de Québec-Université Laval Research Centre, Québec City, QC, Canada
- Department of Medicine, Laval University, Québec City, QC, Canada
| | - André Tchernof
- Endocrinology and Nephrology Unit, CHU de Québec-Université Laval Research Centre, Québec City, QC, Canada
- Department of Medicine, Laval University, Québec City, QC, Canada
- Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Denis Richard
- Department of Medicine, Laval University, Québec City, QC, Canada
- Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
| | - Laurent Biertho
- Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
- Department of Surgery, Laval University, Québec City, QC, Canada
| | - Stefane Lebel
- Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
- Department of Surgery, Laval University, Québec City, QC, Canada
| | - Simon Marceau
- Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada
- Department of Surgery, Laval University, Québec City, QC, Canada
| | - Mathieu Ferron
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Claudia Gagnon
- Endocrinology and Nephrology Unit, CHU de Québec-Université Laval Research Centre, Québec City, QC, Canada.
- Department of Medicine, Laval University, Québec City, QC, Canada.
- Québec Heart and Lung Institute Research Centre, Québec City, QC, Canada.
- Institute of Nutrition and Functional Foods, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
33
|
Liu Z, Sun F, Liu Z, Wang X, Jin M, Mao J, Wu Q, Yan S, Xu K, Wang K, Hu S. Effect of Sleeve Gastrectomy on Glycometabolism via Forkhead Box O1 (FoxO1)/Lipocalin-2 (LCN2) Pathway. Med Sci Monit 2020; 26:e927458. [PMID: 32845875 PMCID: PMC7780888 DOI: 10.12659/msm.927458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The mechanism by which sleeve gastrectomy (SG) improves glycometabolism has remained unclear so far. Increasing evidence has demonstrated that bone is a regulator of glucose metabolism, and osteoblast-derived forkhead box O1 (FoxO1) and lipocalin-2 (LCN2) are regulators of energy metabolism. The aim of this study was to investigate whether the FOXO1/LCN2 signaling pathway is involved in the anti-diabetic effect of SG. MATERIAL AND METHODS Insulin resistance was induced in Wistar rats, which were then intraperitoneally injected with streptozotocin to induce a type 2 diabetic state. Levels of fasting blood glucose, serum insulin, HbA1c, and LCN2 were analyzed at corresponding time points after SG and sham surgeries. The expressions of FOXO1, LCN2, and the melanocortin 4 receptor (MC4R) in bone and hypothalamus were detected by immunofluorescence. FOXO1 siRNA was applied to downregulate FOXO1 expression in osteoblasts of rats. The influence of FOXO1 gene on expression of LCN2 was investigated in cultured osteoblasts by western blot and PCR. RESULTS Glucose metabolism in the SG group was significantly improved. The LCN2 expression in bone in the SG group was higher than that in the sham group, whereas FOXO1 expression in the SG group was lower than that in the sham group. The binding rate of LCN2 and MC4R in the hypothalamus was also higher in the SG group compared with that in the sham group. The downregulation of FOXO1 expression in osteoblasts was accompanied by upregulation of LCN2 expression. CONCLUSIONS These results suggest that the FOXO1/LCN2 signaling pathway participates in the anti-diabetic effect of SG.
Collapse
Affiliation(s)
- Zhi Liu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Fuyun Sun
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Zitian Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaoyang Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Mingxin Jin
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Jiajia Mao
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Qunzheng Wu
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Shaohua Yan
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Kai Xu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Kexin Wang
- Department of General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| | - Sanyuan Hu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
34
|
Ceylan Hİ, Saygın Ö, Özel Türkcü Ü. Assessment of acute aerobic exercise in the morning versus evening on asprosin, spexin, lipocalin-2, and insulin level in overweight/obese versus normal weight adult men. Chronobiol Int 2020; 37:1252-1268. [PMID: 32741294 DOI: 10.1080/07420528.2020.1792482] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of this study was to examine the acute effect of aerobic exercise when performed in the morning and evening on obesity-related hormones of asprosin, spexin, lipocalin-2, and insulin in normal weight (NW) and overweight/obese (OW/OO) adults. A total of 20 adult male individuals (10 NW and 10 OW/OO) volunteered their participation. Both groups were subjected to an aerobic exercise protocol in moderate intensity (heart rate reserve of 55-59%) for 30 min at two different time periods of the day (morning: 08:00-10:00 h, evening: 20.00-22.00 h) at least 3 d apart. BeBis analysis revealed the OW/OO group consumed significantly less energy (1781.59 ± 410.71 kcal) as compared with NW group (2380.28 ± 445.50 kcal) before the evening exercise (about 3 d) (p <.05). As compared with the NW group, basal serum asprosin, insulin, and lipocalin-2 hormone levels were higher in the OW/OO group, and serum spexin level was lower in OW/OO group (p <.05). Body temperature significantly increased after morning and evening aerobic exercise in both groups. The increase in body temperature was significantly higher after the evening exercise in the OW/OO group compared to the NW group (p <.05). Significant decrease in serum asprosin lipocalin-2, and insulin levels was observed in both groups after exercise (p <.05). Evening aerobic exercise more greatly decreased serum asprosin, lipocalin-2, and insulin level in the OW/OO group as compared with the NW group (p <.05). In conclusion, it is thought that negative energy balance caused by psychological energy restriction and evening aerobic exercise, which leads to a further increase in body temperature, triggers greater decrease of orexigenic signals (suppression of appetite), and is more effective in the development of adipose tissue inflammation and insulin sensitivity, especially in OW/OO group.
Collapse
Affiliation(s)
- Halil İbrahim Ceylan
- Faculty of Kazim Karabekir Education, Physical Education and Sports Teaching Department, Ataturk University , Erzurum, Turkey
| | - Özcan Saygın
- Faculty of Sports Sciences, Coaching Science Department, Mugla Sitki Kocman University , Muğla, Turkey
| | - Ümmühani Özel Türkcü
- Faculty of Medicine, Medical Biochemistry Department, Mugla Sitki Kocman University , Muğla, Turkey
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe the current state of our thinking regarding bone-muscle interactions beyond the mechanical perspective. RECENT FINDINGS Recent and prior evidence has begun to dissect many of the molecular mechanisms that bone and muscle use to communicate with each other and to modify each other's function. Several signaling factors produced by muscle and bone have emerged as potential mediators of these biochemical/molecular interactions. These include muscle factors such as myostatin, Irisin, BAIBA, IL-6, and the IGF family and the bone factors FGF-23, Wnt1 and Wnt3a, PGE2, FGF9, RANKL, osteocalcin, and sclerostin. The identification of these signaling molecules and their underlying mechanisms offers the very real and exciting possibility that new pharmaceutical approaches can be developed that will permit the simultaneous treatments of diseases that often occur in combination, such as osteoporosis and sarcopenia.
Collapse
Affiliation(s)
- Nuria Lara-Castillo
- Department of Oral and Craniofacial Sciences, UMKC School of Dentistry, 650 East 25th Street, Kansas City, MO, 64108, USA
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, UMKC School of Dentistry, 650 East 25th Street, Kansas City, MO, 64108, USA.
| |
Collapse
|
36
|
Banjabi AA, Li AJ, Kumosani TA, Yousef JM, Kannan K. Serum concentrations of perfluoroalkyl substances and their association with osteoporosis in a population in Jeddah, Saudi Arabia. ENVIRONMENTAL RESEARCH 2020; 187:109676. [PMID: 32485360 DOI: 10.1016/j.envres.2020.109676] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 06/11/2023]
Abstract
Human exposure to poly- and perfluoroalkyl substances (PFASs) is widespread and has received considerable attention in recent years due to their link with adverse health outcomes, including bone health. Nevertheless, no earlier studies have reported serum PFAS concentrations, and their association with incident osteoporosis in populations in Saudi Arabia. In this clinical case-control study, serum samples collected from 208 individuals (n = 100 cases and n = 108 controls) aged 40-89 years from Jeddah, Saudi Arabia, were analyzed for 17 PFASs. Unconditional logistic regression was used to calculate odds ratios (ORs) for association between serum PFAS concentrations and osteoporosis, stratified by gender, age, serum calcium and vitamin D, previous history of fractures and thyroid disorders. Perfluorooctanoic acid (PFOA), perfluorooctane sulfonic acid (PFOS), perfluorobutanoic acid (PFBA), perfluorononanoic acid (PFNA), perfluorohexane sulfonic acid (PFHxS), perfluoro-n-pentanoic acid (PFPeA) and perfluoroundecanoic acid (PFUnDA) were detected in >80% of serum samples analyzed. PFOS (overall median concentration: 5.08 ng/mL), PFHxS (1.49 ng/mL), PFOA (1.33 ng/mL) and PFNA (0.55 ng/mL) accounted for 94% and 80% of the total serum PFASs concentrations in cases and controls, respectively. Serum PFOA, PFNA and PFUnDA concentrations increased with age in Saudi women. Results from the crude models showed that individuals in the 2nd, 3rd and 4th quartiles of serum PFAS concentrations had 2.3-96-fold increase in odds of diagnosis for osteoporosis compared with those in the 1st quartile (rank order: PFUnDA > PFOA > PFNA > PFOS > PFHxS). Our results suggest that exposure to PFOA, PFOS, PFNA, PFHxS and PFUnDA was associated with osteoporosis in this sample of adult Saudi population.
Collapse
Affiliation(s)
- Abeer A Banjabi
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adela Jing Li
- Wadsworth Center, New York State Department of Health, Albany, NY, 12201, USA
| | - Taha A Kumosani
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Production of Bio-products for Industrial Applications Research Group, King Abdulaziz University, Jeddah, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jehad M Yousef
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Biochemistry Department, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Kurunthachalam Kannan
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Wadsworth Center, New York State Department of Health, Albany, NY, 12201, USA; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Pediatrics, New York University School of Medicine, New York, NY, 10016, USA; Department of Environmental Medicine, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
37
|
Graham ZA, Liu XH, Harlow L, Pan J, Azulai D, Tawfeek HA, Wnek RD, Mattingly AJ, Bauman WA, Yarrow JF, Cardozo CP. Effects of a High-Fat Diet on Tissue Mass, Bone, and Glucose Tolerance after Chronic Complete Spinal Cord Transection in Male Mice. Neurotrauma Rep 2020; 1:17-31. [PMID: 34223527 PMCID: PMC8240892 DOI: 10.1089/neur.2020.0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is associated with obesity and is a risk factor for type 2 diabetes mellitus (T2DM). Immobilization, muscle atrophy, obesity, and loss of sympathetic innervation to the liver are believed to contribute to risks of these abnormalities. Systematic study of the mechanisms underlying SCI-induced metabolic disorders has been limited by a lack of animal models of insulin resistance following SCI. Therefore, the effects of a high-fat diet (HFD), which causes weight gain and glucose intolerance in neurologically intact mice, was tested in mice that had undergone a spinal cord transection at thoracic vertebra 10 (T10) or a sham-transection. At 84 days after surgery, Sham-HFD and SCI-HFD mice showed impaired intraperitoneal glucose tolerance when compared with Sham control (Sham-Con) or SCI control (SCI-Con) mice fed a standard control chow. Glucose tolerance in SCI-Con mice was comparable to that of Sham-Con mice. The mass of paralyzed skeletal muscle, liver, and epididymal, inguinal, and omental fat deposits were lower in SCI versus Sham groups, with lower liver mass present in SCI-HFD versus SCI-Con animals. SCI also produced sublesional bone loss, with no differences between SCI-Con and SCI-HFD groups. The results suggest that administration of a HFD to mice after SCI may provide a model to better understand mechanisms leading to insulin resistance post-SCI, as well as an approach to study pathogenesis of glucose intolerance that is independent of obesity.
Collapse
Affiliation(s)
- Zachary A Graham
- Research Service, Birmingham VA Medical Center, Birmingham, Alabama, USA.,Department of Cell, Developmental and Integrative Biology, University of Alabama-Birmingham, Birmingham, Alabama, USA
| | - Xin-Hua Liu
- Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Harlow
- Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Jiangping Pan
- Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Daniella Azulai
- Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Hesham A Tawfeek
- Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Russell D Wnek
- Research Service and Brain Rehabilitation Research Center, Malcolm Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, Florida, USA
| | - Alex J Mattingly
- Research Service and Brain Rehabilitation Research Center, Malcolm Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, Florida, USA
| | - William A Bauman
- Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua F Yarrow
- Research Service and Brain Rehabilitation Research Center, Malcolm Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, Florida, USA.,Division of Endocrinology, Diabetes, and Metabolism, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Christopher P Cardozo
- Center for the Medical Consequences of SCI, James J. Peters VA Medical Center, Bronx, NY, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
38
|
Crosstalk of Brain and Bone-Clinical Observations and Their Molecular Bases. Int J Mol Sci 2020; 21:ijms21144946. [PMID: 32668736 PMCID: PMC7404044 DOI: 10.3390/ijms21144946] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/06/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
As brain and bone disorders represent major health issues worldwide, substantial clinical investigations demonstrated a bidirectional crosstalk on several levels, mechanistically linking both apparently unrelated organs. While multiple stress, mood and neurodegenerative brain disorders are associated with osteoporosis, rare genetic skeletal diseases display impaired brain development and function. Along with brain and bone pathologies, particularly trauma events highlight the strong interaction of both organs. This review summarizes clinical and experimental observations reported for the crosstalk of brain and bone, followed by a detailed overview of their molecular bases. While brain-derived molecules affecting bone include central regulators, transmitters of the sympathetic, parasympathetic and sensory nervous system, bone-derived mediators altering brain function are released from bone cells and the bone marrow. Although the main pathways of the brain-bone crosstalk remain ‘efferent’, signaling from brain to bone, this review emphasizes the emergence of bone as a crucial ‘afferent’ regulator of cerebral development, function and pathophysiology. Therefore, unraveling the physiological and pathological bases of brain-bone interactions revealed promising pharmacologic targets and novel treatment strategies promoting concurrent brain and bone recovery.
Collapse
|
39
|
Gonzalez-Gil AM, Elizondo-Montemayor L. The Role of Exercise in the Interplay between Myokines, Hepatokines, Osteokines, Adipokines, and Modulation of Inflammation for Energy Substrate Redistribution and Fat Mass Loss: A Review. Nutrients 2020; 12:E1899. [PMID: 32604889 PMCID: PMC7353393 DOI: 10.3390/nu12061899] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022] Open
Abstract
Exercise is an effective strategy for preventing and treating obesity and its related cardiometabolic disorders, resulting in significant loss of body fat mass, white adipose tissue browning, redistribution of energy substrates, optimization of global energy expenditure, enhancement of hypothalamic circuits that control appetite-satiety and energy expenditure, and decreased systemic inflammation and insulin resistance. Novel exercise-inducible soluble factors, including myokines, hepatokines, and osteokines, and immune cytokines and adipokines are hypothesized to play an important role in the body's response to exercise. To our knowledge, no review has provided a comprehensive integrative overview of these novel molecular players and the mechanisms involved in the redistribution of metabolic fuel during and after exercise, the loss of weight and fat mass, and reduced inflammation. In this review, we explain the potential role of these exercise-inducible factors, namely myokines, such as irisin, IL-6, IL-15, METRNL, BAIBA, and myostatin, and hepatokines, in particular selenoprotein P, fetuin A, FGF21, ANGPTL4, and follistatin. We also describe the function of osteokines, specifically osteocalcin, and of adipokines such as leptin, adiponectin, and resistin. We also emphasize an integrative overview of the pleiotropic mechanisms, the metabolic pathways, and the inter-organ crosstalk involved in energy expenditure, fat mass loss, reduced inflammation, and healthy weight induced by exercise.
Collapse
Affiliation(s)
- Adrian M. Gonzalez-Gil
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
- Tecnologico de Monterrey, Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, San Pedro Garza Garcia P.C. 66278, Mexico
| |
Collapse
|
40
|
Jørgensen CV, Bräuner‐Osborne H. Pharmacology and physiological function of the orphan GPRC6A receptor. Basic Clin Pharmacol Toxicol 2020; 126 Suppl 6:77-87. [DOI: 10.1111/bcpt.13397] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Christinna V. Jørgensen
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Hans Bräuner‐Osborne
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
41
|
Lacombe J, Al Rifai O, Loter L, Moran T, Turcotte AF, Grenier-Larouche T, Tchernof A, Biertho L, Carpentier AC, Prud'homme D, Rabasa-Lhoret R, Karsenty G, Gagnon C, Jiang W, Ferron M. Measurement of bioactive osteocalcin in humans using a novel immunoassay reveals association with glucose metabolism and β-cell function. Am J Physiol Endocrinol Metab 2020; 318:E381-E391. [PMID: 31935114 PMCID: PMC7395472 DOI: 10.1152/ajpendo.00321.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Osteocalcin (OCN) is a bone-derived hormone involved in the regulation of glucose metabolism. In serum, OCN exists in carboxylated and uncarboxylated forms (ucOCN), and studies in rodents suggest that ucOCN is the bioactive form of this hormone. Whether this is also the case in humans is unclear, because a reliable assay to measure ucOCN is not available. Here, we established and validated a new immunoassay (ELISA) measuring human ucOCN and used it to determine the level of bioactive OCN in two cohorts of overweight or obese subjects, with or without type 2 diabetes (T2D). The ELISA could specifically detect ucOCN concentrations ranging from 0.037 to 1.8 ng/mL. In a first cohort of overweight or obese postmenopausal women without diabetes (n = 132), ucOCN correlated negatively with fasting glucose (r = -0.18, P = 0.042) and insulin resistance assessed by the homeostatic model assessment of insulin resistance (r = -0.18, P = 0.038) and positively with insulin sensitivity assessed by a hyperinsulinemic-euglycemic clamp (r = 0.18, P = 0.043) or insulin sensitivity index derived from an oral glucose tolerance test (r = 0.26, P = 0.003). In a second cohort of subjects with severe obesity (n = 16), ucOCN was found to be lower in subjects with T2D compared with those without T2D (2.76 ± 0.38 versus 4.52 ± 0.06 ng/mL, P = 0.009) and to negatively correlate with fasting glucose (r = -0.50, P = 0.046) and glycated hemoglobin (r = -0.57, P = 0.021). Moreover, the subjects with ucOCN levels below 3 ng/mL had a reduced insulin secretion rate during a hyperglycemic clamp (P = 0.03). In conclusion, ucOCN measured with this novel and specific assay is inversely associated with insulin resistance and β-cell dysfunction in humans.
Collapse
Affiliation(s)
- Julie Lacombe
- Unité de Recherche en Physiologie Moléculaire, Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
| | - Omar Al Rifai
- Unité de Recherche en Physiologie Moléculaire, Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Québec, Canada
| | | | - Thomas Moran
- Center for Therapeutic Antibody Development, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anne-Frédérique Turcotte
- Endocrinology and Nephrology Unit, CHU de Québec-Université Laval Research Center, Québec City, Québec, Canada
| | - Thomas Grenier-Larouche
- Québec Heart and Lung Institute Research Centre, Québec City, Québec, Canada
- Service d'Endocrinologie, Département de Médecine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - André Tchernof
- Québec Heart and Lung Institute Research Centre, Québec City, Québec, Canada
| | - Laurent Biertho
- Québec Heart and Lung Institute Research Centre, Québec City, Québec, Canada
| | - André C Carpentier
- Service d'Endocrinologie, Département de Médecine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Denis Prud'homme
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
- Institut du Savoir Montfort, Ottawa, Ontario, Canada
| | - Rémi Rabasa-Lhoret
- Département de Nutrition, Université de Montréal, Montréal, Québec, Canada
- Unité de Recherche en Maladies Métaboliques, Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, New York
| | - Claudia Gagnon
- Endocrinology and Nephrology Unit, CHU de Québec-Université Laval Research Center, Québec City, Québec, Canada
- Québec Heart and Lung Institute Research Centre, Québec City, Québec, Canada
- Department of Medicine, Université Laval, Québec City, Québec, Canada
| | | | - Mathieu Ferron
- Unité de Recherche en Physiologie Moléculaire, Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Québec, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| |
Collapse
|
42
|
Cipriani C, Colangelo L, Santori R, Renella M, Mastrantonio M, Minisola S, Pepe J. The Interplay Between Bone and Glucose Metabolism. Front Endocrinol (Lausanne) 2020; 11:122. [PMID: 32265831 PMCID: PMC7105593 DOI: 10.3389/fendo.2020.00122] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 02/24/2020] [Indexed: 12/13/2022] Open
Abstract
The multiple endocrine functions of bone other than those related to mineral metabolism, such as regulation of insulin sensitivity, glucose homeostasis, and energy metabolism, have recently been discovered. In vitro and murine studies investigated the impact of several molecules derived from osteoblasts and osteocytes on glucose metabolism. In addition, the effect of glucose on bone cells suggested a mutual cross-talk between bone and glucose homeostasis. In humans, these mechanisms are the pivotal determinant of the skeletal fragility associated with both type 1 and type 2 diabetes. Metabolic abnormalities associated with diabetes, such as increase in adipose tissue, reduction of lean mass, effects of hyperglycemia per se, production of the advanced glycation end products, diabetes-associated chronic kidney disease, and perturbation of the calcium-PTH-vitamin D metabolism, are the main mechanisms involved. Finally, there have been multiple reports of antidiabetic drugs affecting the skeleton, with differences among basic and clinical research data, as well as of anti-osteoporosis medication influencing glucose metabolism. This review focuses on the aspects linking glucose and bone metabolism by offering insight into the most recent evidence in humans.
Collapse
|
43
|
Abstract
Abstract
The bones form the framework of our body. We know that bones protect our vital organs, regulate calcium and phosphorous homeostasis, and function as a site of erythropoiesis. More recently, however, the identification of bone hormones has allowed us to envision bones as endocrine organs too. Within the last few years, the bone hormones osteocalcin and lipocalin 2 have been implicated with glucose and energy metabolism. We systematically reviewed articles surrounding this subject and found a clear relationship between the osteocalcin levels and glucose tolerance and insulin sensitivity. We also found that many journals have shown the detrimental effects of an absences of lipocalin 2 from adipocytes. As osteocalcin administration to mice showed decreased blood glucose levels and promoted glucose tolerance and insulin sensitivity. Future studies could perhaps explore the use of osteocalcin as a supplement for type 2 diabetes.
Collapse
|
44
|
Abstract
Osteoblasts are specialized mesenchymal cells that synthesize bone matrix and coordinate the mineralization of the skeleton. These cells work in harmony with osteoclasts, which resorb bone, in a continuous cycle that occurs throughout life. The unique function of osteoblasts requires substantial amounts of energy production, particularly during states of new bone formation and remodelling. Over the last 15 years, studies have shown that osteoblasts secrete endocrine factors that integrate the metabolic requirements of bone formation with global energy balance through the regulation of insulin production, feeding behaviour and adipose tissue metabolism. In this article, we summarize the current understanding of three osteoblast-derived metabolic hormones (osteocalcin, lipocalin and sclerostin) and the clinical evidence that suggests the relevance of these pathways in humans, while also discussing the necessity of specific energy substrates (glucose, fatty acids and amino acids) to fuel bone formation and promote osteoblast differentiation.
Collapse
Affiliation(s)
- Naomi Dirckx
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Megan C Moorer
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA.
| |
Collapse
|
45
|
Singh RG, Nguyen NN, Cervantes A, Kim JU, Stuart CE, Petrov MS. Circulating levels of lipocalin-2 are associated with fatty pancreas but not fatty liver. Peptides 2019; 119:170117. [PMID: 31276730 DOI: 10.1016/j.peptides.2019.170117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/12/2019] [Accepted: 06/29/2019] [Indexed: 12/12/2022]
Abstract
Lipocalin-2 (LCN-2), a peptide with diverse expression pattern, has been identified as a biomarker of various diseases as well as a factor contributing to inflammatory responses associated with excess adiposity and ensuing metabolic disorders. Although the inter-relationship between LCN-2 and excess adiposity is increasingly recognized, little is known about the inter-relationship between LCN-2 and ectopic fat deposition. The present study aimed to investigate the associations between LCN-2 and fatty pancreas as well as fatty liver. In addition, the associations between LCN-2 and pro-inflammatory cytokines were studied. Magnetic resonance imaging was used to quantify intra-pancreatic fat deposition and visceral-to-subcutaneous fat volume ratio whereas magnetic resonance spectroscopy was used to quantify liver fat deposition. Fasting venous blood was analyzed for LCN-2, C-C motif chemokine ligand 2, interleukin-6, leptin, tumor necrosis factor-α, glycated hemoglobin, glucose, and insulin. Binary logistic regression and linear regression analyses were conducted. Three statistical models were built to adjust for demographics, comorbidities, levels of glycated hemoglobin, insulin resistance, and abdominal fat distribution. A total of 79 individuals were studied, of whom 20 had fatty pancreas, 14 had fatty liver, and 4 had both. Lipocalin-2 was significantly associated with fatty pancreas in all the adjusted models (p = 0.014 in the most adjusted model) but was not significantly associated with fatty liver in any of the studied models. Lipocalin-2 was significantly associated with interleukin-6 and tumor necrosis factor-α, in both the unadjusted and adjusted models. Leptin and C-C motif chemokine ligand 2 were not significantly associated with LCN-2 in any of the studied models. These findings suggest that LCN-2 is a potential biomarker of fatty pancreas, independent of abdominal fat distribution, insulin resistance, and other covariates. The role of LCN-2 in intra-pancreatic fat deposition and related low-grade inflammation warrants further investigations.
Collapse
Affiliation(s)
- Ruma G Singh
- School of Medicine, University of Auckland, Auckland, New Zealand
| | - Ngoc Nhu Nguyen
- School of Medicine, University of Auckland, Auckland, New Zealand
| | - Aya Cervantes
- School of Medicine, University of Auckland, Auckland, New Zealand
| | - Jin U Kim
- School of Medicine, University of Auckland, Auckland, New Zealand
| | | | - Maxim S Petrov
- School of Medicine, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
46
|
Baldini G, Phelan KD. The melanocortin pathway and control of appetite-progress and therapeutic implications. J Endocrinol 2019; 241:R1-R33. [PMID: 30812013 PMCID: PMC6500576 DOI: 10.1530/joe-18-0596] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/22/2019] [Indexed: 12/19/2022]
Abstract
The initial discovery that ob/ob mice become obese because of a recessive mutation of the leptin gene has been crucial to discover the melanocortin pathway to control appetite. In the melanocortin pathway, the fed state is signaled by abundance of circulating hormones such as leptin and insulin, which bind to receptors expressed at the surface of pro-opiomelanocortin (POMC) neurons to promote processing of POMC to the mature hormone α-melanocyte-stimulating hormone (α-MSH). The α-MSH released by POMC neurons then signals to decrease energy intake by binding to melanocortin-4 receptor (MC4R) expressed by MC4R neurons to the paraventricular nucleus (PVN). Conversely, in the 'starved state' activity of agouti-related neuropeptide (AgRP) and of neuropeptide Y (NPY)-expressing neurons is increased by decreased levels of circulating leptin and insulin and by the orexigenic hormone ghrelin to promote food intake. This initial understanding of the melanocortin pathway has recently been implemented by the description of the complex neuronal circuit that controls the activity of POMC, AgRP/NPY and MC4R neurons and downstream signaling by these neurons. This review summarizes the progress done on the melanocortin pathway and describes how obesity alters this pathway to disrupt energy homeostasis. We also describe progress on how leptin and insulin receptors signal in POMC neurons, how MC4R signals and how altered expression and traffic of MC4R change the acute signaling and desensitization properties of the receptor. We also describe how the discovery of the melanocortin pathway has led to the use of melanocortin agonists to treat obesity derived from genetic disorders.
Collapse
Affiliation(s)
- Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kevin D. Phelan
- Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
47
|
Idelevich A, Baron R. Brain to bone: What is the contribution of the brain to skeletal homeostasis? Bone 2018; 115:31-42. [PMID: 29777919 PMCID: PMC6110971 DOI: 10.1016/j.bone.2018.05.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
The brain, which governs most, if not all, physiological functions in the body, from the complexities of cognition, learning and memory, to the regulation of basal body temperature, heart rate and breathing, has long been known to affect skeletal health. In particular, the hypothalamus - located at the base of the brain in close proximity to the medial eminence, where the blood-brain-barrier is not as tight as in other regions of the brain but rather "leaky", due to fenestrated capillaries - is exposed to a variety of circulating body cues, such as nutrients (glucose, fatty acids, amino acids), and hormones (insulin, glucagon, leptin, adiponectin) [1-3].Information collected from the body via these peripheral cues is integrated by hypothalamic sensing neurons and glial cells [4-7], which express receptors for these nutrients and hormones, transforming these cues into physiological outputs. Interestingly, many of the same molecules, including leptin, adiponectin and insulin, regulate both energy and skeletal homeostasis. Moreover, they act on a common set of hypothalamic nuclei and their residing neurons, activating endocrine and neuronal systems, which ultimately fine-tune the body to new physiological states. This review will focus exclusively on the brain-to-bone pathway, highlighting the most important anatomical sites within the brain, which are known to affect bone, but not covering the input pathways and molecules informing the brain of the energy and bone metabolic status, covered elsewhere [8-10]. The discussion in each section will present side by side the metabolic and bone-related functions of hypothalamic nuclei, in an attempt to answer some of the long-standing questions of whether energy is affected by bone remodeling and homeostasis and vice versa.
Collapse
Affiliation(s)
- Anna Idelevich
- Department of Medicine, Harvard Medical School and Endocrine Unit MGH, Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Roland Baron
- Department of Medicine, Harvard Medical School and Endocrine Unit MGH, Division of Bone and Mineral Metabolism, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
48
|
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: 3.8] [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
|
49
|
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
|
50
|
Emerging and Established Models of Bone Metastasis. Cancers (Basel) 2018; 10:cancers10060176. [PMID: 29865211 PMCID: PMC6024970 DOI: 10.3390/cancers10060176] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/30/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023] Open
Abstract
Metastasis is the leading cause of cancer-related death and drives patient morbidity as well as healthcare costs. Bone is the primary site of metastasis for several cancers—breast and prostate cancers in particular. Efforts to treat bone metastases have been stymied by a lack of models to study the progression, cellular players, and signaling pathways driving bone metastasis. In this review, we examine newly described and classic models of bone metastasis. Through the use of current in vivo, microfluidic, and in silico computational bone metastasis models we may eventually understand how cells escape the primary tumor and how these circulating tumor cells then home to and colonize the bone marrow. Further, future models may uncover how cells enter and then escape dormancy to develop into overt metastases. Recreating the metastatic process will lead to the discovery of therapeutic targets for disrupting and treating bone metastasis.
Collapse
|