1
|
Zhang X, Nguyen MH. Metabolic dysfunction-associated steatotic liver disease: A sexually dimorphic disease and breast and gynecological cancer. Metabolism 2025; 167:156190. [PMID: 40081614 DOI: 10.1016/j.metabol.2025.156190] [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: 01/02/2025] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become a global public health and economic burden worldwide in the past few decades. Epidemiological studies have shown that MASLD is a multisystem disease that is associated not only with liver-related complications but also with an increased risk of developing extrahepatic cancers. MASLD is a sexually dimorphic disease with sex hormones playing an important role in the development and progression of MASLD, especially by the levels and ratios of circulating estrogens and androgens. MASLD is associated with hormone-sensitive cancers including breast and gynecological cancer. The risk of breast and gynecological cancer is elevated in individuals with MASLD driven by shared metabolic risk factors including obesity and insulin resistance. Multiple potential mechanisms underline these associations including metabolic dysfunction, gut dysbiosis, chronic inflammation and dysregulated release of hepatokines. However, the effect of hormone therapy including hormone replacement therapy and anti-estrogen treatment on MASLD and female-specific cancers remains debatable at this time. This synopsis will review the associations between MASLD and breast and gynecological cancer, their underlying mechanisms, implications of hormonal therapies, and their future directions.
Collapse
Affiliation(s)
- Xinrong Zhang
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States; Department of Epidemiology and Population Health, Stanford University Medical Center, Palo Alto, CA, United States; Stanford Cancer Institute, Stanford University Medical Center, Palo Alto, CA, United States.
| |
Collapse
|
2
|
Hu J, Yang Y, Fu S, Yu X, Wang X. Exercise improves glucose and lipid metabolism in high fat diet feeding male mice through androgen/androgen receptor-mediated metabolism regulatory factors. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167926. [PMID: 40419170 DOI: 10.1016/j.bbadis.2025.167926] [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: 11/08/2024] [Revised: 05/22/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
Exercise alleviates high fat diet (HFD)-induced glycolipid metabolism disorders, but the mechanisms are not discovered totally. Low androgen/androgen receptor (AR) levels are associated with glycolipid metabolism disorders in males, and androgen/AR modulate glycolipid metabolism-related regulators such as peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1-α), Forkhead box O1 (FoxO1), phosphoenolpyruvate carboxy kinase (PEPCK) and stearyl-coenzyme A desaturase 1 (SCD1). In the present study, we blockaded androgen (by castration) and inhibited or activated AR activity (by AR antagonist flutamide and agonist S4, respectively) to clarify androgen/AR's roles in exercise-induced alleviation of glycolipid metabolism disorders in male high fat diet (HFD) feeding mice and the underlying mechanisms. We found that: (1) exercise reversed HFD-induced glycolipid metabolism disorders, including restoring fasting blood glucose (FBG), glucose tolerance, TC and TG, accompanied with the increases of serum testosterone and AR in muscle and liver. (2) castration exacerbated HFD-induced impairment of glucose tolerance and increases of TC and TG, and abolished the lowering effect of exercise on FBG and TC. Flutamide further impaired glucose tolerance, increased plasm LDL content, and attenuated exercise-induced improvements of FBG and TG content. (3) exercise reduced the levels of FoxO1 and SCD1, increased PGC-1α in muscle and liver of HFD mice; whereas castration and flutamide reversed exercise-induced improvements of these indicators. Furthermore, S4 rectified the levels of FoxO1, PGC-1α and SCD1 in HFD mice even in absence of androgen. In conclusion, androgen/AR, especially AR, play important roles in alleviating glycolipid metabolism disorders in HFD mice at sedentary and exercise states, which might fulfill through FOXO1, PGC-1α and SCD1.
Collapse
Affiliation(s)
- Jing Hu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yanan Yang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Shaoting Fu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China; Department of Kinesiology, College of Physical Education, Shanghai Normal University, Shanghai, China
| | - Xiaohan Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiaohui Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
3
|
Barsky ST, Monks DA. The role of androgens and global and tissue-specific androgen receptor expression on body composition, exercise adaptation, and performance. Biol Sex Differ 2025; 16:28. [PMID: 40269952 PMCID: PMC12016402 DOI: 10.1186/s13293-025-00707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/23/2025] [Indexed: 04/25/2025] Open
Abstract
Gonadal testosterone stimulates skeletal muscle anabolism and contributes to sexually differentiated adipose distribution through incompletely understood mechanisms. Observations in humans and animal models have indicated a major role for androgen receptor (AR) in mediating sex differences in body composition throughout the lifespan. Traditional surgical, genetic and pharmacological studies have tested systemic actions of circulating androgens, and more recent transgenic approaches have allowed for tests of AR gene function in specific androgen responsive niches contributing to body composition, including: skeletal muscle and surrounding interstitial cells, white and brown adipose, as well as trabecular and cortical bone. Less well understood is how these functions of gonadal androgens interact with exercise. Here, we summarize the understood mechanisms of action of AR and its interactions with exercise, specifically on outcomes of body composition and muscle function, and the global- and tissue-specific role of AR in regulating skeletal muscle, adipose, and bone morphology. Additionally, we describe the known effects of androgen and AR manipulation on female body composition, muscle morphology, and sport performance, while highlighting a need for greater inclusion of female subjects in human and animal muscle physiology and endocrinology research.
Collapse
Affiliation(s)
- Sabrina Tzivia Barsky
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, ON, Canada
| | - Douglas Ashley Monks
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, ON, Canada.
- Department of Psychology, Faculty of Arts & Science, University of Toronto Mississauga, 3359 Mississauga Road North, Deerfield Hall DH4098, Mississauga, ON, L5L 1C6, Canada.
| |
Collapse
|
4
|
Hu S, Kang H, Bae M, Kim MB, Jang H, Corvino O, Pham TX, Lee Y, Smyth JA, Park YK, Lee JY. Histone Deacetylase 9 Deletion Inhibits Hepatic Steatosis and Adipose Tissue Inflammation in Male Diet-Induced Obese Mice. J Gastroenterol Hepatol 2025; 40:741-749. [PMID: 39730208 PMCID: PMC11875955 DOI: 10.1111/jgh.16856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/29/2024]
Abstract
AIM The goal of this study was to determine the role of histone deacetylase 9 (HDAC9) in the development of diet-induced metabolic dysfunction-associated steatohepatitis (MASH) and white adipose tissue (WAT) dysfunctions. METHODS We fed male and female mice with global Hdac9 knockout (KO) and their wild-type (WT) littermates an obesogenic high-fat/high-sucrose/high-cholesterol (35%/34%/2%, w/w) diet for 20 weeks. RESULTS Hdac9 deletion markedly inhibited body weight gain and liver steatosis with lower liver weight and triglyceride content than WT in male mice but not females. Consistently, hepatic expression of genes crucial for de novo lipogenesis was markedly suppressed only in male, but not female, Hdac9 KO mice. However, Hdac9 deletion had a minimal effect on hepatic inflammation and fibrosis. In WAT, Hdac9 KO showed less adipocyte hypertrophy, inflammation, and fibrosis in male mice compared with WT. In addition, indirect calorimetry demonstrated that male Hdac9 KO mice had significantly higher metabolic rates, respiratory exchange ratios, and energy expenditure without altering physical activities than WT, which was not observed in female mice. CONCLUSIONS Our findings indicate that global deletion of Hdac9 prevented the development of obesity, hepatic steatosis, and WAT inflammation and fibrosis in male mice with diet-induced obesity and MASH, suggesting that a sex-dependent role of HDAC9 may exist in the pathways mentioned above.
Collapse
Affiliation(s)
- Siqi Hu
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Hyunju Kang
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
- Department of Food and Nutrition, Keimyung University, Daegu, South Korea
| | - Minkyung Bae
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
- Department of Food and Nutrition, Yonsei University, Seoul, South Korea
| | - Mi-Bo Kim
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Hyungryun Jang
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Olivia Corvino
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Tho X Pham
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Yoojin Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Joan A Smyth
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT 06269, USA
| | - Young-Ki Park
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
5
|
Kim M, Shin D. Effects of the Interaction Between Oxidative Balance Score and Polygenic Risk Scores on Incidence of Metabolic Syndrome in Middle-Aged Korean Adults. Antioxidants (Basel) 2024; 13:1556. [PMID: 39765884 PMCID: PMC11672919 DOI: 10.3390/antiox13121556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/03/2025] Open
Abstract
Oxidative stress is implicated in insulin resistance, obesity, and metabolic syndromes (MetSs). However, the interplay between oxidative stress and genetic predisposition during the development of MetS remains unclear. In this study, we aimed to investigate the effects of the interaction between oxidative balance score (OBS) and polygenic risk score (PRS) on the incidence of MetS in middle-aged Korean adults. We analyzed data from 25,879 participants aged ≥40 years from the Health Examinees Cohort of the Korean Genome and Epidemiology Study. The OBS was calculated using 11 antioxidant and five pro-oxidant factors. A genome-wide association study and clumping analysis identified 16 independent single-nucleotide polymorphisms associated with MetS that were used to calculate individual PRSs. Multivariable Cox proportional hazard models adjusted for confounding variables were used to assess the impact of OBS and PRS on the incidence of MetS. During a mean follow-up period of 4.3 years, we recorded 3153 cases of MetS. In both men and women, the group with the lowest OBS and a high PRS had a 1.50-fold (hazard ratio [HR] 1.50, 95% confidence interval [CI] 1.07-2.11) and 1.89-fold (HR 1.89, 95% CI 1.40-2.56) higher incidence, respectively, of MetS compared to those with the highest OBS and a low PRS. Among women with a high PRS, the HRs decreased significantly across OBS quintiles 1 through 5 (p for trend = 0.009). These findings suggest that managing the oxidative balance may be particularly crucial for individuals with a high genetic risk for MetS.
Collapse
Affiliation(s)
| | - Dayeon Shin
- Department of Food and Nutrition, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea;
| |
Collapse
|
6
|
Forner-Piquer I, Giommi C, Sella F, Lombó M, Montik N, Dalla Valle L, Carnevali O. Endocannabinoid System and Metabolism: The Influences of Sex. Int J Mol Sci 2024; 25:11909. [PMID: 39595979 PMCID: PMC11593739 DOI: 10.3390/ijms252211909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
The endocannabinoid system (ECS) is a lipid signaling system involved in numerous physiological processes, such as endocrine homeostasis, appetite control, energy balance, and metabolism. The ECS comprises endocannabinoids, their cognate receptors, and the enzymatic machinery that tightly regulates their levels within tissues. This system has been identified in various organs, including the brain and liver, in multiple mammalian and non-mammalian species. However, information regarding the sex-specific regulation of the ECS remains limited, even though increasing evidence suggests that interactions between sex steroid hormones and the ECS may ultimately modulate hepatic metabolism and energy homeostasis. Within this framework, we will review the sexual dimorphism of the ECS in various animal models, providing evidence of the crosstalk between endocannabinoids and sex hormones via different metabolic pathways. Additionally, we will underscore the importance of understanding how endocrine-disrupting chemicals and exogenous cannabinoids influence ECS-dependent metabolic pathways in a sex-specific manner.
Collapse
Affiliation(s)
- Isabel Forner-Piquer
- Centre for Pollution Research and Policy, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK;
| | - Christian Giommi
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (C.G.); (F.S.)
- INBB—Biostructures and Biosystems National Institute, 00136 Roma, Italy
| | - Fiorenza Sella
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (C.G.); (F.S.)
- INBB—Biostructures and Biosystems National Institute, 00136 Roma, Italy
| | - Marta Lombó
- INBB—Biostructures and Biosystems National Institute, 00136 Roma, Italy
- Department of Molecular Biology, Universidad de León, 24071 León, Spain
| | - Nina Montik
- Department of Odontostomatological and Specialized Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy;
| | | | - Oliana Carnevali
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (C.G.); (F.S.)
- INBB—Biostructures and Biosystems National Institute, 00136 Roma, Italy
| |
Collapse
|
7
|
Sakai H, Uno H, Yamakawa H, Tanaka K, Ikedo A, Uezumi A, Ohkawa Y, Imai Y. The androgen receptor in mesenchymal progenitors regulates skeletal muscle mass via Igf1 expression in male mice. Proc Natl Acad Sci U S A 2024; 121:e2407768121. [PMID: 39292748 PMCID: PMC11441553 DOI: 10.1073/pnas.2407768121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024] Open
Abstract
Androgens exert their effects primarily by binding to the androgen receptor (AR), a ligand-dependent nuclear receptor. While androgens have anabolic effects on skeletal muscle, previous studies reported that AR functions in myofibers to regulate skeletal muscle quality, rather than skeletal muscle mass. Therefore, the anabolic effects of androgens are exerted via nonmyofiber cells. In this context, the cellular and molecular mechanisms of AR in mesenchymal progenitors, which play a crucial role in maintaining skeletal muscle homeostasis, remain largely unknown. In this study, we demonstrated expression of AR in mesenchymal progenitors and found that targeted AR ablation in mesenchymal progenitors reduced limb muscle mass in mature adult, but not young or aged, male mice, although fatty infiltration of muscle was not affected. The absence of AR in mesenchymal progenitors led to remarkable perineal muscle hypotrophy, regardless of age, due to abnormal regulation of transcripts associated with cell death and extracellular matrix organization. Additionally, we revealed that AR in mesenchymal progenitors regulates the expression of insulin-like growth factor 1 (Igf1) and that IGF1 administration prevents perineal muscle atrophy in a paracrine manner. These findings indicate that the anabolic effects of androgens regulate skeletal muscle mass via, at least in part, AR signaling in mesenchymal progenitors.
Collapse
Affiliation(s)
- Hiroshi Sakai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| | - Hideaki Uno
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| | - Harumi Yamakawa
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka812-0054, Japan
| | - Aoi Ikedo
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime791-0295, Japan
| | - Akiyoshi Uezumi
- Division of Cell Heterogeneity, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka812-0054, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka812-0054, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime791-0295, Japan
| |
Collapse
|
8
|
Kositanurit W, Siritaweechai N, Varachotisate P, Burana C, Sukswai N, Surintrspanont J, Siriviriyakul P, Kaikaew K, Werawatganon D. Genistein mitigates diet-induced obesity and metabolic dysfunctions in gonadectomized mice with some sex-differential effects. Front Endocrinol (Lausanne) 2024; 15:1392866. [PMID: 39351533 PMCID: PMC11439649 DOI: 10.3389/fendo.2024.1392866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Background Obesity is associated with insulin resistance (IR) and metabolic dysfunction-associated steatotic liver disease (MASLD). Genistein, an isoflavone, is a promising natural compound for preventing and treating obesity and metabolic dysfunctions. We aimed to investigate the sex-specific protective effects of genistein on obesity, IR, and MASLD in a murine model of sex hormone deprivation with diet-induced obesity (DIO), mimicking postmenopausal women or aging men with metabolic syndrome. Methods Gonadectomized and sham-operated C57BL/6NJcl mice were fed a high-fat high-sucrose diet for 4 weeks to induce obesity (7 mice per group). In gonadectomized mice, genistein (16 mg/kg/day) or vehicle (7.5% dimethyl sulfoxide) was orally administered for 45 days. We assessed glucose homeostasis parameters, hepatic histopathology, and hepatic gene expression to investigate the effects of gonadectomy and genistein treatment. Results Gonadectomy exacerbated adiposity in both sexes. Ovariectomy diminished the protective effects of female gonadal hormones on the homeostatic model assessment for insulin resistance (HOMA-IR), serum alanine transaminase levels, hepatic steatosis score, and the expression of hepatic genes associated with MASLD progression and IR, such as Fasn, Srebf1, Saa1, Cd36, Col1a1, Pck1, and Ppargc1a. Genistein treatment in gonadectomized mice significantly reduced body weight gain and the hepatic steatosis score in both sexes. However, genistein treatment significantly attenuated HOMA-IR and the expression of the hepatic genes only in female mice. Conclusion Genistein treatment mitigates DIO-related MASLD in both male and female gonadectomized mice. Regarding hepatic gene expression associated with MASLD and IR, the beneficial effect of genistein was significantly evident only in female mice. This study suggests a potential alternative application of genistein in individuals with obesity and sex hormone deprivation, yet pending clinical trials.
Collapse
Affiliation(s)
- Weerapat Kositanurit
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natakorn Siritaweechai
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pachara Varachotisate
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chuti Burana
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Narittee Sukswai
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jerasit Surintrspanont
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pathology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Prasong Siriviriyakul
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Kasiphak Kaikaew
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Duangporn Werawatganon
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
9
|
Prapaharan B, Lea M, Beaudry JL. Weighing in on the role of brown adipose tissue for treatment of obesity. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13157. [PMID: 39087083 PMCID: PMC11290130 DOI: 10.3389/jpps.2024.13157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Brown adipose tissue (BAT) activation is an emerging target for obesity treatments due to its thermogenic properties stemming from its ability to shuttle energy through uncoupling protein 1 (Ucp1). Recent rodent studies show how BAT and white adipose tissue (WAT) activity can be modulated to increase the expression of thermogenic proteins. Consequently, these alterations enable organisms to endure cold-temperatures and elevate energy expenditure, thereby promoting weight loss. In humans, BAT is less abundant in obese subjects and impacts of thermogenesis are less pronounced, bringing into question whether energy expending properties of BAT seen in rodents can be translated to human models. Our review will discuss pharmacological, hormonal, bioactive, sex-specific and environmental activators and inhibitors of BAT to determine the potential for BAT to act as a therapeutic strategy. We aim to address the feasibility of utilizing BAT modulators for weight reduction in obese individuals, as recent studies suggest that BAT's contributions to energy expenditure along with Ucp1-dependent and -independent pathways may or may not rectify energy imbalance characteristic of obesity.
Collapse
Affiliation(s)
| | | | - Jacqueline L. Beaudry
- Temerty Faculty of Medicine, Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Booijink R, Ramachandran P, Bansal R. Implications of innate immune sexual dimorphism for MASLD pathogenesis and treatment. Trends Pharmacol Sci 2024; 45:614-627. [PMID: 38853100 DOI: 10.1016/j.tips.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Growing evidence suggests that metabolic dysfunction-associated steatotic liver disease (MASLD) is significantly higher in men versus women. Increased prevalence is observed in postmenopausal women, suggesting that age and sex (hormones) influence MASLD development and progression. Molecular data further reveal that sex regulates the innate immune responses with an essential role in MASLD progression. To date, there has been limited focus on the role of innate immune sexual dimorphism in MASLD, and differences between men and women are not considered in the current drug discovery landscape. In this review, we summarize the sex disparities and innate immune sexual dimorphism in MASLD pathogenesis. We further highlight the importance of harnessing sexual dimorphism in identifying therapeutic targets, developing pharmacological therapies, and designing (pre-) clinical studies for the personalized treatment for MASLD.
Collapse
Affiliation(s)
- Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Prakash Ramachandran
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, Edinburgh, UK
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
11
|
Du M, Qiu M, Qian Y, Wang T, Chen X. Serum Copper/Zinc Ratio in Overweight and Obese Children: a Cross-Sectional Study. Biol Trace Elem Res 2024; 202:1539-1549. [PMID: 37505435 DOI: 10.1007/s12011-023-03790-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
To assess the association between serum copper/zinc ratio (CZR) and overweight/obesity in children. Sociodemographic and clinical data of children aged 8-18 years old were extracted from the National Health and Nutrition Examination Survey (NHANES) database in this cross-sectional study. These associations were explored using weighted univariate and multivariate logistic regression analyses with the odds ratios (ORs) and 95% confidence intervals (CIs). High serum CZR level was related to higher odds of overweight (OR = 1.74), obesity (OR = 5.26), and central obesity (OR = 2.99). Subgroup analyses showed that high serum CZR levels were associated with high odds of overweight in children aged ≥ 12 years old (OR = 2.19) and females (OR = 2.02), while the increased odds of obesity and central obesity were found in children aged ≥ 12 years old and both male and female. Elevated serum CZR level was linked to high odds of childhood overweight, obesity, and central obesity, and this relationship was also found in children with different age and gender.
Collapse
Affiliation(s)
- Mu Du
- Department of Endocrinology, Children's Hospital of Capital Institute of Pediatrics, No. 2 Yabao Road, Chaoyang District, 100020, Beijing, People's Republic of China
| | - Mingfang Qiu
- Department of Endocrinology, Children's Hospital of Capital Institute of Pediatrics, No. 2 Yabao Road, Chaoyang District, 100020, Beijing, People's Republic of China
| | - Ye Qian
- Department of Endocrinology, Children's Hospital of Capital Institute of Pediatrics, No. 2 Yabao Road, Chaoyang District, 100020, Beijing, People's Republic of China
| | - Tianqi Wang
- Department of Endocrinology, Children's Hospital of Capital Institute of Pediatrics, No. 2 Yabao Road, Chaoyang District, 100020, Beijing, People's Republic of China
| | - Xiaobo Chen
- Department of Endocrinology, Children's Hospital of Capital Institute of Pediatrics, No. 2 Yabao Road, Chaoyang District, 100020, Beijing, People's Republic of China.
| |
Collapse
|
12
|
Hu B, Zhou W, Deng X, Sun M, Sun R, Li Q, Ren J, Jiang W, Wang Y, Liu S, Zhan J. Structural analysis of polysaccharide from Inonotus obliquus and investigate combined impact on the sex hormones, intestinal microbiota and metabolism in SPF male mice. Int J Biol Macromol 2024; 262:129686. [PMID: 38331071 DOI: 10.1016/j.ijbiomac.2024.129686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/04/2024] [Accepted: 01/21/2024] [Indexed: 02/10/2024]
Abstract
The dysregulation of sex hormone levels is associated with metabolic disorders such as obesity. Inonotus obliquus polysaccharide (IOP) exhibits a promising therapeutic effect on conditions like obesity and diabetes, potentially linked to its influence on intestinal microbiota and metabolism. The exact cause and mechanisms that link sex hormones, gut microbiota and metabolism are still unknown. In this research, we examined the molecular weight, monosaccharide composition, and glycosidic bond type of IOP. We found that IOP mostly consists of alpha-structured 6‑carbon glucopyranose, with a predominant (1 → 4) linkage to monosaccharides and a uniform distribution. Following this, we administered two different concentrations of IOP to mice through gavage. The results of the enzyme-linked immunosorbent assay (ELISA) demonstrated a significant increase in testosterone (T) levels in the IOP group as compared to the control group. Additionally, the results of tissue immunofluorescence indicated that increased IOP led to a decrease in adiponectin content and an increase in SET protein expression. The study also revealed changes in the intestinal microbiota and metabolic changes in mice through 16S rRNA data and non-targeted LC-MS data, respectively. The study also found that IOP mainly affects pathways linked to glycerophospholipid metabolism. In addition, it has been observed that there is an increase in the number of beneficial bacteria, such as the Eubacterium coprostanoligenes group and g.Lachnospiraceae NK4A136 group, while the levels of metabolites that are linked to obesity or diabetes, such as 1,5-anhydrosorbitol, are reduced. Furthermore, biomarker screening has revealed that the main microorganism responsible for the differences between the three groups is g.Erysipelatoclostridiaceae. In summary, these findings suggest that IOP exerts its therapeutic effects through a synergistic interplay between sex hormones, gut microbiome composition, and metabolic processes.
Collapse
Affiliation(s)
- Binhong Hu
- College of Chemistry and life Sciences, Chengdu Normal University, China; Department of Forest Mycology and Plant pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden; Sichuan Provincial key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, Sichuan, China.
| | - Wenjing Zhou
- College of Chemistry and life Sciences, Chengdu Normal University, China; College of Veterinary Medicine, Yangzhou University (Institute of Comparative Medicine), Yangzhou, China
| | - Xin Deng
- College of Chemistry and life Sciences, Chengdu Normal University, China; College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Mengxue Sun
- College of Chemistry and life Sciences, Chengdu Normal University, China
| | - Rong Sun
- College of Chemistry and life Sciences, Chengdu Normal University, China
| | - Qing Li
- College of Chemistry and life Sciences, Chengdu Normal University, China
| | - Jingyuan Ren
- College of Chemistry and life Sciences, Chengdu Normal University, China
| | - Wei Jiang
- College of Chemistry and life Sciences, Chengdu Normal University, China; Sichuan Provincial key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, Sichuan, China
| | - Yanping Wang
- College of Chemistry and life Sciences, Chengdu Normal University, China; Sichuan Provincial key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, Sichuan, China
| | - Songqing Liu
- College of Chemistry and life Sciences, Chengdu Normal University, China; Sichuan Provincial key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, Sichuan, China
| | - Jiasui Zhan
- Department of Forest Mycology and Plant pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| |
Collapse
|
13
|
Chen XJ. Analysis of Sex Hormones, Insulin Dosage, and Risk Factors Associated With Male Diabetic Patients. Am J Mens Health 2024; 18:15579883241235062. [PMID: 38554046 PMCID: PMC10981860 DOI: 10.1177/15579883241235062] [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: 09/13/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 04/01/2024] Open
Abstract
The purpose of this study is to assess the influence of sex hormones and other indicators on risk factors for hypercoagulable states in male patients with type 2 diabetes mellitus. Ninety-two diabetic patients were divided into two groups based on testosterone levels: T1 group (testosterone <12 mmol/L) and T2 group (testosterone >12 mmol/L). Fifty-four non-diabetic patients were used as the control group. Sex hormone index, glucose, insulin, C-peptide, 24-h urinary free cortisol, thromboelastography, and insulin resistance index were measured by radioimmunoassay. Testosterone was lower in the diabetic men than in the control group (12.02 vs 14.77, p < .05), and was inversely related to blood coagulation status, blood glucose level, and cortisol level. Body mass index was positively correlated with estradiol and insulin resistance index. Testosterone was independently associated with the clotting process after controlling for age. Low testosterone is a risk factor for hypercoagulable state in diabetic men. Elevated estradiol and insulin resistance are influential factors for increased body mass index.
Collapse
Affiliation(s)
- Xiao-Jun Chen
- Department of Endocrinology, Chongming Branch, Shanghai Tenth People’s Hospital, Shanghai, China
| |
Collapse
|
14
|
Chen Z, Zhang E, Gan L, Jiang G, Duan Q, Huang M, Li H, Huang G. Analysis of the association between testosterone and cardiovascular disease potential risk factor apolipoprotein B in adult males without cancer: national health and nutrition examination survey 2011-2016. Front Endocrinol (Lausanne) 2024; 15:1304344. [PMID: 38435750 PMCID: PMC10905265 DOI: 10.3389/fendo.2024.1304344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Background Over the years, there has been extensive exploration of the association between testosterone and lipid profiles, yet the precise mechanisms underlying their interaction remain incompletely elucidated. Similarly, there is a dearth of research on the correlation between serum apolipoprotein B (apoB) and serum total testosterone (TT), particularly within specific populations. Methods We conducted a cross-sectional study to assess the relationship between serum TT concentration and serum apoB concentration. Using the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2016, we employed weighted generalized linear models, weighted univariate, weighted multivariate analysis, and smooth curve fitting to assist in exploring the relationship between serum TT and apoB. Serum apoB concentration served as the independent variable, and serum TT concentration as the dependent variable. ApoB was divided into four quartiles-Q1 (<0.7g/L, N=691), Q2 (≥0.7g/L to <0.9g/L, N=710), Q3 (≥0.9g/L to <1.1g/L, N=696), and Q4 (≥1.1g/L, N=708)-thereby further solidifying the stable association between the two. Additionally, the application of smooth curve fitting will contribute to a more detailed elucidation of the specific relationship between serum TT concentration and serum apoB concentration under different factors (Drinking, Smoke, Diabetes, Hypertension, and High cholesterol level.). Results The results indicate a negative correlation between serum TT concentration and apoB concentration (β=-113.4; 95% CI: -146.6, -80.2; P<0.001). After adjusting for confounding variables, the negative correlation between apoB concentration and TT concentration remains significant (β=-61.0; 95% CI: -116.7, -5.2; P=0.040). When apoB concentration was converted from a continuous variable to a categorical variable (quartiles: Q1<0.7g/L; Q2:≥0.7g/L to<0.9g/L; Q3:≥0.9g/L to <1.1g/L; Q4: ≥1.1g/L), TT level of participants in the highest quartile (≥1.1g/L) was -47.2 pg/mL (95% CI: -91.2, -3.3; P=0.045) lower than that in the lowest quartile (<0.7g/L). The smooth curve fitting diagram revealed differences in the relationship between TT concentration and apoB among individuals with different cardiovascular disease (CVD) risk factors. Conclusions This study elucidates a robust inverse correlation between serum TT concentration and apoB concentration, maintaining statistical significance even upon adjustment for confounding factors. These findings present a promising avenue for addressing the prevention and treatment of low testosterone and CVD.
Collapse
Affiliation(s)
- Zhiyi Chen
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Enpu Zhang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Lu Gan
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Ganggang Jiang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Qilin Duan
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Mou Huang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Huizhen Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Guixiao Huang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| |
Collapse
|
15
|
Barsky ST, Monks DA. Lifespan Effects of Muscle-Specific Androgen Receptor Overexpression on Body Composition of Male and Female Rats. Endocrinology 2024; 165:bqae012. [PMID: 38301268 DOI: 10.1210/endocr/bqae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/03/2024]
Abstract
Androgenic actions of gonadal testosterone are thought to be a major mechanism promoting sex differences in body composition across the lifespan. However, this inference is based on studies of androgen receptor (AR) function in late adolescent or emerging adult rodents. Here we assess body composition and AR expression in skeletal muscle of rats at defined ages, comparing wild-type (WT) to transgenic human skeletal actin-driven AR overexpression (HSAAR) rats which overexpress AR in skeletal muscle. Male and female HSAAR and WT Sprague Dawley rats (N = 288) underwent dual-energy x-ray absorptiometry (DXA) scanning and tissue collection at postnatal day (PND) 1, 10, 21, 42, 70, 183, 243, and 365. Expected sex differences in body composition and muscle mass largely onset with puberty (PND-21), with no associated changes to skeletal muscle AR protein. In adulthood, HSAAR increased tibialis anterior (TA) and extensor digitorum longus mass in males, and reduced the expected gain in gonadal fat mass in both sexes. In WT rats, AR protein was reduced in soleus, but not TA, throughout life. Nonetheless, soleus AR protein expression was greater in male rats than female rats at all ages of sexual development, yet only at PND-70 in TA. Overall, despite muscle AR overexpression effects, results are inconsistent with major sex differences in body composition during sexual development being driven by changes in muscle AR, rather suggesting that changes in ligand promote sexual differentiation of body composition during pubertal timing. Nonetheless, increased skeletal muscle AR in adulthood can be sufficient to increase muscle mass in males, and reduce adipose in both sexes.
Collapse
Affiliation(s)
- Sabrina Tzivia Barsky
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Douglas Ashley Monks
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario M5S 3G5, Canada
- Department of Psychology, Faculty of Arts & Science, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| |
Collapse
|
16
|
Hutchison AL, Tavaglione F, Romeo S, Charlton M. Endocrine aspects of metabolic dysfunction-associated steatotic liver disease (MASLD): Beyond insulin resistance. J Hepatol 2023; 79:1524-1541. [PMID: 37730124 DOI: 10.1016/j.jhep.2023.08.030] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023]
Abstract
While the association of metabolic dysfunction-associated steatotic liver disease (MASLD) with obesity and insulin resistance is widely appreciated, there are a host of complex interactions between the liver and other endocrine axes. While it can be difficult to definitively distinguish direct causal relationships and those attributable to increased adipocyte mass, there is substantial evidence of the direct and indirect effects of endocrine dysregulation on the severity of MASLD, with strong evidence that low levels of growth hormone, sex hormones, and thyroid hormone promote the development and progression of disease. The impact of steroid hormones, e.g. cortisol and dehydroepiandrosterone, and adipokines is much more divergent. Thoughtful assessment, based on individual risk factors and findings, and management of non-insulin endocrine axes is essential in the evaluation and management of MASLD. Multiple therapeutic options have emerged that leverage various endocrine axes to reduce the fibroinflammatory cascade in MASH.
Collapse
Affiliation(s)
| | - Federica Tavaglione
- Clinical Medicine and Hepatology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy; Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Cardiology Department, Sahlgrenska University Hospital, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Michael Charlton
- Center for Liver Diseases, University of Chicago, United States.
| |
Collapse
|
17
|
Lawande NV, Conklin EA, Christian‐Hinman CA. Sex and gonadectomy modify behavioral seizure susceptibility and mortality in a repeated low-dose kainic acid systemic injection paradigm in mice. Epilepsia Open 2023; 8:1512-1522. [PMID: 37715318 PMCID: PMC10690657 DOI: 10.1002/epi4.12828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVE Sex differences in epilepsy appear driven in part due to effects of gonadal steroids, with varying results in experimental models based on species, strain, and method of seizure induction. Furthermore, removing the main source of these steroids via gonadectomy may impact seizure characteristics differently in males and females. Repeated low-dose kainic acid (RLDKA) systemic injection paradigms were recently shown to reliably induce status epilepticus (SE) and hippocampal histopathology in C57BL/6J mice. Here, we investigated whether seizure susceptibility in a RLDKA injection protocol exhibits a sex difference and whether gonadectomy differentially influences response to this seizure induction paradigm in males and females. METHODS Adult C57BL/6J mice were left gonad-intact as controls or gonadectomized (females: ovariectomized, OVX; males: orchidectomized, ORX). At least 2 weeks later, KA was injected ip, every 30 minutes at 7.5 mg/kg or less until the animal reached SE, defined by at least 5 generalized seizures (GS, Racine stage 3 or higher). Parameters of susceptibility to GS induction, SE development, and mortality rates were quantified. RESULTS No differences in seizure susceptibility or mortality were observed between control males and control females. Gonadectomized mice exhibited increased susceptibility and reduced latency to both GS and SE in comparison to corresponding controls of the same sex, but the effects were stronger in males. In addition, ORX males, but not OVX females, exhibited strongly increased seizure-induced mortality. SIGNIFICANCE The RLDKA protocol is notable for its efficacy in inducing SE and seizure-induced histopathology in C57BL/6J mice, the background for many transgenic strains in current use in epilepsy research. The present results indicate that this protocol may be beneficial for investigating the effects of gonadal hormone replacement on seizure susceptibility, mortality, and seizure-induced histopathology, and that gonadectomy unmasks sex differences in susceptibility to seizures and mortality not observed in gonad-intact controls.
Collapse
Affiliation(s)
- Niraj V. Lawande
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | | | - Catherine A. Christian‐Hinman
- Department of Molecular and Integrative PhysiologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
18
|
Rizk J, Sahu R, Duteil D. An overview on androgen-mediated actions in skeletal muscle and adipose tissue. Steroids 2023; 199:109306. [PMID: 37634653 DOI: 10.1016/j.steroids.2023.109306] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Androgens are a class of steroid hormones primarily associated with male sexual development and physiology, but exert pleiotropic effects in either sex. They have a crucial role in various physiological processes, including the regulation of skeletal muscle and adipose tissue homeostasis. The effects of androgens are mainly mediated through the androgen receptor (AR), a ligand-activated nuclear receptor expressed in both tissues. In skeletal muscle, androgens via AR exert a multitude of effects, ranging from increased muscle mass and strength, to the regulation of muscle fiber type composition, contraction and metabolic functions. In adipose tissue, androgens influence several processes including proliferation, fat distribution, and metabolism but they display depot-specific and organism-specific effects which differ in certain context. This review further explores the potential mechanisms underlying androgen-AR signaling in skeletal muscle and adipose tissue. Understanding the roles of androgens and their receptor in skeletal muscle and adipose tissue is essential for elucidating their contributions to physiological processes, disease conditions, and potential therapeutic interventions.
Collapse
Affiliation(s)
- Joe Rizk
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
| | - Rajesh Sahu
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
| | - Delphine Duteil
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France.
| |
Collapse
|
19
|
Dubois V, Ciancia S, Doms S, El Kharraz S, Sommers V, Kim NR, David K, Van Dijck J, Valle-Tenney R, Maes C, Antonio L, Decallonne B, Carmeliet G, Claessens F, Cools M, Vanderschueren D. Testosterone Restores Body Composition, Bone Mass, and Bone Strength Following Early Puberty Suppression in a Mouse Model Mimicking the Clinical Strategy in Trans Boys. J Bone Miner Res 2023; 38:1497-1508. [PMID: 37222072 DOI: 10.1002/jbmr.4832] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 05/13/2023] [Indexed: 05/25/2023]
Abstract
Transgender youth increasingly present at pediatric gender services. Some of them receive long-term puberty suppression with gonadotropin-releasing hormone analogues (GnRHa) before starting gender-affirming hormones (GAH). The impact of GnRHa use started in early puberty on bone composition and bone mass accrual is unexplored. It is furthermore unclear whether subsequent GAH fully restore GnRHa effects and whether the timing of GAH introduction matters. To answer these questions, we developed a mouse model mimicking the clinical strategy applied in trans boys. Prepubertal 4-week-old female mice were treated with GnRHa alone or with GnRHa supplemented with testosterone (T) from 6 weeks (early puberty) or 8 weeks (late puberty) onward. Outcomes were analyzed at 16 weeks and compared with untreated mice of both sexes. GnRHa markedly increased total body fat mass, decreased lean body mass, and had a modest negative impact on grip strength. Both early and late T administration shaped body composition to adult male levels, whereas grip strength was restored to female values. GnRHa-treated animals showed lower trabecular bone volume and reduced cortical bone mass and strength. These changes were reversed by T to female levels (cortical bone mass and strength) irrespective of the time of administration or even fully up to adult male control values (trabecular parameters) in case of earlier T start. The lower bone mass in GnRHa-treated mice was associated with increased bone marrow adiposity, also reversed by T. In conclusion, prolonged GnRHa use started in prepubertal female mice modifies body composition toward more fat and less lean mass and impairs bone mass acquisition and strength. Subsequent T administration counteracts GnRHa impact on these parameters, shaping body composition and trabecular parameters to male values while restoring cortical bone architecture and strength up to female but not male control levels. These findings could help guide clinical strategies in transgender care. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Vanessa Dubois
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
- Basic and Translational Endocrinology (BaTE), Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Silvia Ciancia
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Stefanie Doms
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
| | - Sarah El Kharraz
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Vera Sommers
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Na Ri Kim
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
| | - Karel David
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Jolien Van Dijck
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Roger Valle-Tenney
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Leen Antonio
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Martine Cools
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Pediatric Endocrinology Service, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (Chrometa), KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Kumar A, Narkar VA. Nuclear receptors as potential therapeutic targets in peripheral arterial disease and related myopathy. FEBS J 2023; 290:4596-4613. [PMID: 35942640 PMCID: PMC9908775 DOI: 10.1111/febs.16593] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 12/31/2022]
Abstract
Peripheral arterial disease (PAD) is a prevalent cardiovascular complication of limb vascular insufficiency, causing ischemic injury, mitochondrial metabolic damage and functional impairment in the skeletal muscle, and ultimately leading to immobility and mortality. While potential therapies have been mostly focussed on revascularization, none of the currently available pharmacological treatments are fully effective in PAD, often leading to amputations, particularly in chronic metabolic diseases. One major limitation of focussed angiogenesis and revascularization as a therapeutic strategy is a limited effect on metabolic restoration and muscle regeneration in the affected limb. Therefore, additional preclinical investigations are needed to discover novel treatment options for PAD preferably targeting multiple aspects of muscle recovery. In this review, we propose nuclear receptors expressed in the skeletal muscle as potential candidates for ischemic muscle repair in PAD. We review classic steroid and orphan receptors that have been reported to be involved in the regulation of paracrine muscle angiogenesis, oxidative metabolism, mitochondrial biogenesis and muscle regeneration, and discuss how these receptors could be critical for recovery from ischemic muscle damage. Furthermore, we identify existing gaps in our understanding of nuclear receptor signalling in the skeletal muscle and propose future areas of research that could be instrumental in exploring nuclear receptors as therapeutic candidates for treating PAD.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, UTHealth McGovern Medical School, Houston, TX, 77030
- University of Texas MD Anderson and UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030
| |
Collapse
|
21
|
Barsky ST, Monks DA. Androgen action on myogenesis throughout the lifespan; comparison with neurogenesis. Front Neuroendocrinol 2023; 71:101101. [PMID: 37669703 DOI: 10.1016/j.yfrne.2023.101101] [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: 04/28/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/07/2023]
Abstract
Androgens' pleiotropic actions in promoting sex differences present not only a challenge to providing a comprehensive account of their function, but also an opportunity to gain insights by comparing androgenic actions across organ systems. Although often overlooked by neuroscientists, skeletal muscle is another androgen-responsive organ system which shares with the nervous system properties of electrochemical excitability, behavioral relevance, and remarkable capacity for adaptive plasticity. Here we review androgenic regulation of mitogenic plasticity in skeletal muscle with the goal of identifying areas of interest to those researching androgenic mechanisms mediating sexual differentiation of neurogenesis. We use an organizational-activational framework to relate broad areas of similarity and difference between androgen effects on mitogenesis in muscle and brain throughout the lifespan, from early organogenesis, through pubertal organization, adult activation, and aging. The focus of the review is androgenic regulation of muscle-specific stem cells (satellite cells), which share with neural stem cells essential functions in development, plasticity, and repair, albeit with distinct, muscle-specific features. Also considered are areas of paracrine and endocrine interaction between androgen action on muscle and nervous system, including mediation of neural plasticity of innervating and distal neural populations by muscle-produced trophic factors.
Collapse
Affiliation(s)
- Sabrina Tzivia Barsky
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario, Canada.
| | - Douglas Ashley Monks
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, Faculty of Arts & Science, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| |
Collapse
|
22
|
Maes B, Fayazpour F, Catrysse L, Lornet G, Van De Velde E, De Wolf C, De Prijck S, Van Moorleghem J, Vanheerswynghels M, Deswarte K, Descamps B, Vanhove C, Van der Schueren B, Vangoitsenhoven R, Hammad H, Janssens S, Lambrecht BN. STE20 kinase TAOK3 regulates type 2 immunity and metabolism in obesity. J Exp Med 2023; 220:e20210788. [PMID: 37347461 PMCID: PMC10287548 DOI: 10.1084/jem.20210788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 03/31/2023] [Accepted: 06/02/2023] [Indexed: 06/23/2023] Open
Abstract
Healthy adipose tissue (AT) contains ST2+ Tregs, ILC2s, and alternatively activated macrophages that are lost in mice or humans on high caloric diet. Understanding how this form of type 2 immunity is regulated could improve treatment of obesity. The STE20 kinase Thousand And One amino acid Kinase-3 (TAOK3) has been linked to obesity in mice and humans, but its precise function is unknown. We found that ST2+ Tregs are upregulated in visceral epididymal white AT (eWAT) of Taok3-/- mice, dependent on IL-33 and the kinase activity of TAOK3. Upon high fat diet feeding, metabolic dysfunction was attenuated in Taok3-/- mice. ST2+ Tregs disappeared from eWAT in obese wild-type mice, but this was not the case in Taok3-/- mice. Mechanistically, AT Taok3-/- Tregs were intrinsically more responsive to IL-33, through higher expression of ST2, and expressed more PPARγ and type 2 cytokines. Thus, TAOK3 inhibits adipose tissue Tregs and regulates immunometabolism under excessive caloric intake.
Collapse
Affiliation(s)
- Bastiaan Maes
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Farzaneh Fayazpour
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Leen Catrysse
- Cellular and Molecular (Patho)Physiology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Guillaume Lornet
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Evelien Van De Velde
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Caroline De Wolf
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sofie De Prijck
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Justine Van Moorleghem
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Manon Vanheerswynghels
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Kim Deswarte
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Benedicte Descamps
- Department of Electronics and Information Systems, IBiTech-MEDISIP-Infinity Lab, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Department of Electronics and Information Systems, IBiTech-MEDISIP-Infinity Lab, Ghent University, Ghent, Belgium
| | - Bart Van der Schueren
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Roman Vangoitsenhoven
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Bart N. Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam Netherlands
| |
Collapse
|
23
|
Pais R, Cariou B, Noureddin M, Francque S, Schattenberg JM, Abdelmalek MF, Lalazar G, Varma S, Dietrich J, Miller V, Sanyal A, Ratziu V. A proposal from the liver forum for the management of comorbidities in non-alcoholic steatohepatitis therapeutic trials. J Hepatol 2023; 79:829-841. [PMID: 37001695 DOI: 10.1016/j.jhep.2023.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 02/08/2023] [Accepted: 03/13/2023] [Indexed: 06/19/2023]
Abstract
The current document has been developed by the Liver Forum who mandated the NAFLD-Associated Comorbidities Working Group - a multistakeholder group comprised of experts from academic medicine, industry and patient associations - to identify aspects of diverse comorbidities frequently associated with non-alcoholic steatohepatitis (NASH) that can interfere with the conduct of therapeutic trials and, in particular, impact efficacy and safety results. The objective of this paper is to propose guidance for the management of relevant comorbidities in both candidates and actual participants in NASH therapeutic trials. We relied on specific guidelines from scientific societies, when available, but adapted them to the particulars of NASH trials with the aim of addressing multiple interacting requirements such as maintaining patient safety, reaching holistic therapeutic objectives, minimising confounding effects on efficacy and safety of investigational agents and allowing for trial completion. We divided the field of action into: first, analysis and stabilisation of the patient's condition before inclusion in the trial and, second, management of comorbidities during trial conduct. For the former, we discussed the concept of acceptable vs. optimal control of comorbidities, defined metabolic and ponderal stability prior to randomisation and weighed the pros and cons of a run-in period. For the latter, we analysed non-hepatological comorbid conditions for changes or acute events possibly occurring during the trial, including changes in alcohol consumption, in order to detail when specific interventions are necessary and how best to manage concomitant drug intake in line with methodological constraints. These recommendations are intended to act as a guide for clinical trialists and are open to further refinement when additional data become available.
Collapse
Affiliation(s)
- Raluca Pais
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition, France; Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du Thorax, F-44000 Nantes, France
| | | | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Drie Eikenstraat 655, B-2650 Edegem, Belgium; InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Belgium
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC, USA
| | - Gadi Lalazar
- Liver Unit, Digestive Disease Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Sharat Varma
- Novo Nordisk A/S, Vandtårnsvej 108-110, 2860 Søborg Denmark
| | - Julie Dietrich
- GENFIT, Parc Eurasanté 885, Avenue Eugène Avinée, 59120, Loos, France
| | - Veronica Miller
- Forum for Collaborative Research, University of California Berkeley School of Public Health, Washington D.C., USA
| | - Arun Sanyal
- Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Vlad Ratziu
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition, France; INSERM UMRS 1138 CRC, Paris, France.
| |
Collapse
|
24
|
Gielen E, Dupont J, Dejaeger M, Laurent MR. Sarcopenia, osteoporosis and frailty. Metabolism 2023; 145:155638. [PMID: 37348597 DOI: 10.1016/j.metabol.2023.155638] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/21/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023]
Abstract
Muscles and bones are intricately connected tissues displaying marked co-variation during development, growth, aging, and in many diseases. While the diagnosis and treatment of osteoporosis are well established in clinical practice, sarcopenia has only been classified internationally as a disease in 2016. Both conditions are associated with an increased risk of adverse health outcomes such as fractures, dysmobility and mortality. Rather than focusing on one dimension of bone or muscle mass or weakness, the concept of musculoskeletal frailty captures the overall loss of physiological reserves in the locomotor system with age. The term osteosarcopenia in particular refers to the double jeopardy of osteoporosis and sarcopenia. Muscle-bone interactions at the biomechanical, cellular, paracrine, endocrine, neuronal or nutritional level may contribute to the pathophysiology of osteosarcopenia. The paradigm wherein muscle force controls bone strength is increasingly facing competition from a model centering on the exchange of myokines, osteokines and adipokines. The most promising results have been obtained in preclinical models where common drug targets have been identified to treat these conditions simultaneously. In this narrative review, we critically summarize the current understanding of the definitions, epidemiology, pathophysiology, and treatment of osteosarcopenia as part of an integrative approach to musculoskeletal frailty.
Collapse
Affiliation(s)
- Evelien Gielen
- Gerontology and Geriatrics Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Jolan Dupont
- Gerontology and Geriatrics Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Marian Dejaeger
- Gerontology and Geriatrics Unit, Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Michaël R Laurent
- Centre for Metabolic Bone Diseases, University Hospitals Leuven, Leuven, Belgium; Geriatrics Department, Imelda Hospital, Bonheiden, Belgium.
| |
Collapse
|
25
|
Dorfman MD, Monfeuga T, Melhorn SJ, Kanter JE, Frey JM, Fasnacht RD, Chandran A, Lala E, Velasco I, Rubinow KB, Meek TH, Schur EA, Bornfeldt KE, Thaler JP. Central androgen action reverses hypothalamic astrogliosis and atherogenic risk factors induced by orchiectomy and high-fat diet feeding in male mice. Am J Physiol Endocrinol Metab 2023; 324:E461-E475. [PMID: 37053049 PMCID: PMC10202485 DOI: 10.1152/ajpendo.00059.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
Hypogonadism in males confers elevated cardiovascular disease (CVD) risk by unknown mechanisms. Recent radiological evidence suggests that low testosterone (T) is associated with mediobasal hypothalamic (MBH) gliosis, a central nervous system (CNS) cellular response linked to metabolic dysfunction. To address mechanisms linking CNS androgen action to CVD risk, we generated a hypogonadal, hyperlipidemic mouse model with orchiectomy (ORX) combined with hepatic PCSK9 overexpression. After 4 wk of high-fat, high-sucrose diet (HFHS) consumption, despite equal body weights and glucose tolerance, androgen-deficient ORX mice had a more atherogenic lipid profile and increased liver and leukocyte inflammatory signaling compared with sham-operated control mice. Along with these early CVD risk indicators, ORX markedly amplified HFHS-induced astrogliosis in the MBH. Transcriptomic analysis further revealed that ORX and high-fat diet feeding induced upregulation of inflammatory pathways and downregulation of metabolic pathways in hypothalamic astrocytes. To interrogate the role of sex steroid signaling in the CNS in cardiometabolic risk and MBH inflammation, central infusion of T and dihydrotestosterone (DHT) was performed on ORX mice. Central DHT prevented MBH astrogliosis and reduced the liver inflammatory signaling and monocytosis induced by HFHS and ORX; T had a partial protective effect. Finally, a cross-sectional study in 41 adult men demonstrated a positive correlation between radiological evidence of MBH gliosis and plasma lipids. These findings demonstrate that T deficiency in combination with a Western-style diet promotes hypothalamic gliosis concomitant with increased atherogenic risk factors and provide supportive evidence for regulation of lipid metabolism and cardiometabolic risk determinants by the CNS action of sex steroids.NEW & NOTEWORTHY This study provides evidence that hypothalamic gliosis is a key early event through which androgen deficiency in combination with a Western-style diet might lead to cardiometabolic dysregulation in males. Furthermore, this work provides the first evidence in humans of a positive association between hypothalamic gliosis and LDL-cholesterol, advancing our knowledge of CNS influences on CVD risk progression.
Collapse
Affiliation(s)
- Mauricio D Dorfman
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| | | | - Susan J Melhorn
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Jenny E Kanter
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Jeremy M Frey
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Rachael D Fasnacht
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| | | | - Emaad Lala
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Inmaculada Velasco
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Katya B Rubinow
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Thomas H Meek
- Novo Nordisk Research Centre Oxford, Oxford, United Kingdom
| | - Ellen A Schur
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of General Internal Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Karin E Bornfeldt
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States
| | - Joshua P Thaler
- UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, United States
| |
Collapse
|
26
|
Zhang R, Pavan E, Ross AB, Deb-Choudhury S, Dixit Y, Mungure TE, Realini CE, Cao M, Farouk MM. Molecular insights into quality and authentication of sheep meat from proteomics and metabolomics. J Proteomics 2023; 276:104836. [PMID: 36764652 DOI: 10.1016/j.jprot.2023.104836] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Sheep meat (encompassing lamb, hogget and mutton) is an important source of animal protein in many countries, with a unique flavour and sensory profile compared to other red meats. Flavour, colour and texture are the key quality attributes contributing to consumer liking of sheep meat. Over the last decades, various factors from 'farm to fork', including production system (e.g., age, breed, feeding regimes, sex, pre-slaughter stress, and carcass suspension), post-mortem manipulation and processing (e.g., electrical stimulation, ageing, packaging types, and chilled and frozen storage) have been identified as influencing different aspects of sheep meat quality. However conventional meat-quality assessment tools are not able to elucidate the underlying mechanisms and pathways for quality variations. Advances in broad-based analytical techniques have offered opportunities to obtain deeper insights into the molecular changes of sheep meat which may become biomarkers for specific variations in quality traits and meat authenticity. This review provides an overview on how omics techniques, especially proteomics (including peptidomics) and metabolomics (including lipidomics and volatilomics) are applied to elucidate the variations in sheep meat quality, mainly in loin muscles, focusing on colour, texture and flavour, and as tools for authentication. SIGNIFICANCE: From this review, we observed that attempts have been made to utilise proteomics and metabolomics techniques on sheep meat products for elucidating pathways of quality variations due to various factors. For instance, the improvement of colour stability and tenderness could be associated with the changes to glycolysis, energy metabolism and endogenous antioxidant capacity. Several studies identify proteolysis as being important, but potentially conflicting for quality as the enhanced proteolysis improves tenderness and flavour, while reducing colour stability. The use of multiple analytical methods e.g., lipidomics, metabolomics, and volatilomics, detects a wider range of flavour precursors (including both water and lipid soluble compounds) that underlie the possible pathways for sheep meat flavour evolution. The technological advancement in omics (e.g., direct analysis-mass spectrometry) could make analysis of the proteins, lipids and metabolites in sheep meat routine, as well as enhance the confidence in quality determination and molecular-based assurance of meat authenticity.
Collapse
Affiliation(s)
- Renyu Zhang
- Food Technology & Processing, AgResearch Ltd, Palmerston North, New Zealand.
| | - Enrique Pavan
- Food Technology & Processing, AgResearch Ltd, Palmerston North, New Zealand; Unidad Integrada Balcarce (FCA, UNMdP - INTA, EEA Balcarce), Ruta 226 km 73.5, CP7620 Balcarce, Argentina
| | - Alastair B Ross
- Proteins and Metabolites, AgResearch Ltd, Lincoln, New Zealand
| | | | - Yash Dixit
- Food informatics, AgResearch Ltd, Palmerston North, New Zealand
| | | | - Carolina E Realini
- Food Technology & Processing, AgResearch Ltd, Palmerston North, New Zealand
| | - Mingshu Cao
- Data Science, AgResearch Ltd, Palmerston North, New Zealand
| | - Mustafa M Farouk
- Food Technology & Processing, AgResearch Ltd, Palmerston North, New Zealand
| |
Collapse
|
27
|
Klappenbach CM, Wang Q, Jensen AL, Glodosky NC, Delevich K. Sex and timing of gonadectomy relative to puberty interact to influence weight, body composition, and feeding behaviors in mice. Horm Behav 2023; 151:105350. [PMID: 36996734 DOI: 10.1016/j.yhbeh.2023.105350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 04/01/2023]
Abstract
Gonadal sex steroids are important regulators of energy balance in adult rodents, and gonadectomy (GDX) has opposing effects on weight gain in sexually mature males and females. Puberty is associated with the emergence of sex differences in weight, body composition, and feeding behaviors, yet the role of gonadal hormones at puberty remains unclear. To address this, we performed GDX or sham surgery in male and female C57Bl/6 mice at postnatal day (P)25 (prepubertal) or P60 (postpubertal) timepoints and measured weight and body composition for 35 days, after which ad libitum and operant food intake was measured using Feeding Experimentation Device 3 (FED3s) in the home cage. Consistent with previous studies, postpubertal GDX caused weight gain in females and weight loss in males and increased adiposity in both sexes. However, prepubertal GDX decreased weight gain and altered body composition across the adolescent transition (P25 to P60) in males but had no effect in females. Despite the varied effects on weight, GDX decreased food intake and motivation for food as assessed in operant tasks regardless of sex or timing of surgery relative to puberty. Our findings indicate that GDX interacts with both sex and age at surgery to influence weight, body composition, and feeding behavior.
Collapse
Affiliation(s)
- Courtney M Klappenbach
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Qing Wang
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Allison L Jensen
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Nicholas C Glodosky
- Department of Psychology Washington State University, Pullman, WA 99164, USA
| | - Kristen Delevich
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
28
|
Sun F, Piao M, Zhang X, Zhang S, Wei Z, Liu L, Bu Y, Xu S, Zhao X, Meng X, Yue M. Multi-Omics Analysis of Transcriptomic and Metabolomics Profiles Reveal the Molecular Regulatory Network of Marbling in Early Castrated Holstein Steers. Animals (Basel) 2022; 12:3398. [PMID: 36496924 PMCID: PMC9736081 DOI: 10.3390/ani12233398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
The intramuscular fat (IMF), or so-called marbling, is known as potential determinant of the high quality beef in China, Korea, and Japan. Of the methods that affect IMF content in cattle, castration is markedly regarded as an effective and economical way to improve the deposition of IMF but with little attention to its multi-omics in early-castrated cattle. The aim of this study was to investigate the liver transcriptome and metabolome of early-castrated Holstein cattle and conduct a comprehensive analysis of two omics associated with the IMF deposition using transcriptomics and untargeted metabolomics under different treatments: non−castrated and slaughtered at 16 months of age (GL16), castrated at birth and slaughtered at 16 months of age (YL16), and castrated at birth and slaughtered at 26 months of age (YL26). The untargeted metabolome was analyzed using ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The transcriptome of the hepatic genes was analyzed to identify marbling-related genes. Using untargeted metabolomics, the main altered metabolic pathways in the liver of cattle, including those for lipid and amino acid metabolism, were detected in the YL16 group relative to the GL16 and YL26 groups. Significant increases in the presence of betaine, alanine, and glycerol 3-phosphate were observed in the YL16 group (p < 0.05), which might have contributed to the improved beef-marbling production. Compared to the GL16 and YL26 groups, significant increases in the presence of glutathione, acetylcarnitine, and riboflavin but decreases in diethanolamine and 2-hydroxyglutarate were identified in YL16 group (p < 0.05), which might have been beneficial to the beef’s enhanced functional quality. The gene expressions of GLI1 and NUF2 were downregulated and that of CYP3A4 was upregulated in the YL16 group; these results were strongly correlated with the alanine, betaine, and leucine, respectively, in the liver of the cattle. In conclusion, implementation of early castration modified the hepatic metabolites and the related biological pathways by regulating the relevant gene expressions, which could represent a better rearing method for production of high marbled and healthier beef products.
Collapse
Affiliation(s)
- Fang Sun
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Minyu Piao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xinyue Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Siqi Zhang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Ziheng Wei
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Li Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Ye Bu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Shanshan Xu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiaochuan Zhao
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiangren Meng
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Mengmeng Yue
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| |
Collapse
|
29
|
Barsky ST, Monks DA. Myocytic androgen receptor overexpression does not affect sex differences in adaptation to chronic endurance exercise. Biol Sex Differ 2022; 13:59. [PMID: 36274144 PMCID: PMC9590152 DOI: 10.1186/s13293-022-00471-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/06/2022] [Indexed: 12/02/2022] Open
Abstract
Muscle-specific androgen receptor (AR) overexpression (HSAAR transgene) in sedentary male rats results in reduced adiposity, increased mitochondrial enzyme activity, and selective increase in Type 2b myofiber size. Here, we tested chronic endurance exercise interactions with this phenotype in both sexes. Across 9 weeks, rats ran 5×/week on motorized running wheels at increasing speeds and durations. Exercise reduced fat mass in all groups, but sex affected endurance exercise outcomes such that absolute lean mass increased only in females and total body mass decreased only in males. Expected sex differences were observed with males exhibiting greater total body and lean mass; absolute and relative fat mass; bone mineral density; extensor digitorum longus (EDL) myofiber size and glycolytic proportion; but lesser Type 2a and Type 1 myosin expression in tibialis anterior. Observed HSAAR outcomes were not altered by sex, with transgenic rats having greater lean mass, Type 2a myosin expression in soleus, and glycolytic myofiber size in EDL. Tibialis AR content was independently affected by sex, HSAAR, and exercise. No sex differences were observed in tibialis AR expression in wild-type rats, although HSAAR males had greater AR content than HSAAR females. We identified a moderate correlation between AR expression and glycolytic myofiber size, but not whole-body composition. Overall, results suggest myocytic AR overexpression and chronic exercise, despite sharing a similar phenotype to adaptation, are mediated by distinct mechanisms. Further, this study illustrates sex differences in adaptation to chronic endurance exercise, and suggests sex-similarity in the relationship between muscle AR and exercise response. Adaptations in bone, lean, and total mass after forced endurance exercise are sex-dependent in rats. Sex differences in muscle fiber-type size and proportion, lean body mass, and bone density are independent of exercise in rats. Myocytic AR overexpression promotes lean body mass and glycolytic myofiber size in both sexes. Skeletal muscle AR protein is elevated by chronic endurance exercise in rats, and these changes in AR content are correlated with improved glycolytic myofiber size.
Collapse
|
30
|
Chen KW, Chen YS, Chen PJ, Yeh SH. Androgen receptor functions in pericentral hepatocytes to decrease gluconeogenesis and avoid hyperglycemia and obesity in male mice. Metabolism 2022; 135:155269. [PMID: 35914621 DOI: 10.1016/j.metabol.2022.155269] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 07/13/2022] [Accepted: 07/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although the impact of hepatic androgen receptor (AR) pathway on liver pathogenesis was documented, its physiological function in normal liver is remained unclear. This study aims to investigate if hepatic AR acts on metabolism, the major liver function, using a hepatic-specific AR-transgenic (H-ARTG) mouse model. METHODS We established the albumin promoter driven H-ARTG mice and included wild type (WT) and H-ARKO mice for study. The body weight, specific metabolic parameters and results from various tolerance tests were compared in different groups of mice fed a chow diet, from 2 to 18 months of age. Glucose feeding and insulin treatment were used to study the expression and zonal distribution pattern of AR and related genes in liver at different prandial stages. RESULTS The body weight of H-ARTG mice fed a chow diet was 15 % lower than that of wild-type mice, preceded by lower blood glucose and liver triglyceride levels caused by AR reduced hepatic gluconeogenesis. The opposite phenotypes identified in H-ARKO and castrated H-ARTG mice support the critical role of activated AR in decreasing gluconeogenesis and triglyceride levels in liver. Hepatic AR acting by enhancing the expression of cytosolic glycerol-3-phosphate dehydrogenase (cGPDH), a key of glycerophosphate shuttle, was identified as one mechanism to decrease gluconeogenesis from glycerol. We further found AR normally expressed in zone 3 of hepatic lobules. Its level fluctuates dependent on the demand of glucose, decreased by fasting but increased by glucose uptake or insulin stimulation. CONCLUSION AR is a newly identified zone 3 hepatic gene with function in reducing blood glucose and body weight in mice. It suggests that stabilization of hepatic AR is a new direction to prevent hyperglycemia, obesity and nonalcoholic fatty liver disease (NAFLD) in males.
Collapse
Affiliation(s)
- Kai-Wei Chen
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Shan Chen
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Jer Chen
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan; NTU Centers of Genomic and Precision Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shiou-Hwei Yeh
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan; NTU Centers of Genomic and Precision Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
31
|
Takahashi K, Kitaoka Y, Hatta H. Effects of endurance training on metabolic enzyme activity and transporter protein levels in the skeletal muscles of orchiectomized mice. J Physiol Sci 2022; 72:14. [PMID: 35768774 PMCID: PMC10717707 DOI: 10.1186/s12576-022-00839-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/18/2022] [Indexed: 12/20/2022]
Abstract
This study investigated whether endurance training attenuates orchiectomy (ORX)-induced metabolic alterations. At 7 days of recovery after sham operation or ORX surgery, the mice were randomized to remain sedentary or undergo 5 weeks of treadmill running training (15-20 m/min, 60 min, 5 days/week). ORX decreased glycogen concentration in the gastrocnemius muscle, enhanced phosphofructokinase activity in the plantaris muscle, and decreased lactate dehydrogenase activity in the plantaris and soleus muscles. Mitochondrial enzyme activities and protein content in the plantaris and soleus muscles were also decreased after ORX, but preserved, in part, by endurance training. In the treadmill running test (15 m/min, 60 min) after 4 weeks of training, orchiectomized sedentary mice showed impaired exercise performance, which was restored by endurance training. Thus, endurance training could be a potential therapeutic strategy to prevent the hypoandrogenism-induced decline in muscle mitochondrial content and physical performance.
Collapse
Affiliation(s)
- Kenya Takahashi
- Department of Sports Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
| | - Yu Kitaoka
- Department of Human Sciences, Kanagawa University, 3-27-1, Rokkakubashi, Kanagawa-ku, Yokohama, Kanagawa, 221-8686, Japan
| | - Hideo Hatta
- Department of Sports Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| |
Collapse
|
32
|
Lei M, Qu X, Dai Z, Chen R, Zhu H, Shi Z. Effects of Caponization on Growth Performance and Carcass Composition of Yangzhou Ganders. Animals (Basel) 2022; 12:ani12111364. [PMID: 35681829 PMCID: PMC9179501 DOI: 10.3390/ani12111364] [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: 04/14/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Goose meat is recognized as one of the healthiest foods. Goose capons are specially bred and consumed in several parts of China for their high-quality meat. However, the effects of caponization on goose growth and carcass traits have remained uninvestigated, and its molecular mechanisms remain unclear. In this research, caponization lowered testosterone and increased the total cholesterol and triglyceride concentrations in serum. Caponization increased live weights by promoting food intake and abdominal fat deposition, and improved meat quality by increasing intermuscular fat. Changes in the expression of these genes indicate that caponization increases the live weight mainly by increasing fat deposition rather than muscle growth. These results expand our understanding of the mechanisms of caponization on growth performance and fat deposition in ganders. Abstract In this study, we determined the effects of caponization on the growth performance and carcass traits of Yangzhou ganders. Fifty sham operated geese (the control group) and 80 caponized geese (the caponized group) were selected at 150 days of age and reared until 240 days of age. At 210 days of age, 30 geese from the caponized group were selected and fed with testosterone propionate (testosterone group). The results showed that caponization lowered testosterone and increased the total cholesterol and triglyceride concentrations in serum, live weights, average 15 day gains, and feed intake. Abdominal fat and intramuscular fat were significantly higher in the caponized geese than in the control at 240 days. Gene expression analysis showed that caponization promoted abdominal fat deposition and intermuscular fat content by upregulating the expression of adipogenic genes in the liver, adipose tissue, and muscle tissue. The high expression of SOCS3 in the hypothalamus, liver, and muscle of caponized geese suggests that caponization may lead to negative feedback regulation and leptin resistance. Changes in the expression of these genes, along with the downregulation of PAX3 in the breast muscle and MYOG in the leg muscles, indicate that caponization increases the live weight mainly by increasing fat deposition rather than muscle growth. These results expand our understanding of the mechanisms of caponization on growth performance and fat deposition in ganders.
Collapse
Affiliation(s)
- Mingming Lei
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (X.Q.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Nanjing 210014, China
| | - Xiaolu Qu
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (X.Q.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Nanjing 210014, China
| | - Zichun Dai
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (X.Q.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Nanjing 210014, China
| | - Rong Chen
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (X.Q.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Nanjing 210014, China
| | - Huanxi Zhu
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (X.Q.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Nanjing 210014, China
- Correspondence: (H.Z.); (Z.S.); Tel.: +86-025-8439-0346 (H.Z.); +86-025-8439-0956 (Z.S.)
| | - Zhendan Shi
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (X.Q.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Nanjing 210014, China
- Correspondence: (H.Z.); (Z.S.); Tel.: +86-025-8439-0346 (H.Z.); +86-025-8439-0956 (Z.S.)
| |
Collapse
|
33
|
Varghese M, Griffin C, Abrishami S, Eter L, Lanzetta N, Hak L, Clemente J, Agarwal D, Lerner A, Westerhoff M, Patel R, Bowers E, Islam M, Subbaiah P, Singer K. Sex hormones regulate metainflammation in diet-induced obesity in mice. J Biol Chem 2021; 297:101229. [PMID: 34599964 PMCID: PMC8526779 DOI: 10.1016/j.jbc.2021.101229] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/13/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
Men have a statistically higher risk of metabolic and cardiovascular disease than premenopausal women, but the mechanisms mediating these differences are elusive. Chronic inflammation during obesity contributes to disease risk and is significantly more robust in males. Prior work demonstrated that compared with obese males, obese females have reduced proinflammatory adipose tissue macrophages (ATMs). Given the paucity of data on how sex hormones contribute to macrophage responses in obesity, we sought to understand the role of sex hormones in promoting obesity-induced myeloid inflammation. We used gonadectomy, estrogen receptor-deficient alpha chimeras, and androgen-insensitive mice to model sex hormone deficiency. These models were evaluated in diet-induced obesity conditions (high-fat diet [HFD]) and in vitro myeloid assays. We found that ovariectomy increased weight gain and adiposity. Ovariectomized females had increased ATMs and bone marrow myeloid colonies compared with sham-gonadectomized females. In addition, castrated males exposed to HFD had improved glucose tolerance, insulin sensitivity, and adiposity with fewer Ly6chi monocytes and bone marrow myeloid colonies compared with sham-gonadectomized males, although local adipose inflammation was enhanced. Similar findings were observed in androgen-insensitive mice; however, these mice had fewer CD11c+ ATMs, implying a developmental role for androgens in myelopoiesis and adipose inflammation. We concluded that gonadectomy results in convergence of metabolic and inflammatory responses to HFD between the sexes, and that myeloid estrogen receptor alpha contributes minimally to diet-induced inflammatory responses, whereas loss of androgen-receptor signaling improves metabolic and inflammatory outcomes. These studies demonstrate that sex hormones play a critical role in sex differences in obesity, metabolic dysfunction, and myeloid inflammation.
Collapse
Affiliation(s)
- Mita Varghese
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cameron Griffin
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Simin Abrishami
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Leila Eter
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas Lanzetta
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Layla Hak
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeremy Clemente
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Devyani Agarwal
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Arianna Lerner
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Maria Westerhoff
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ravi Patel
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Emily Bowers
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mohammed Islam
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Perla Subbaiah
- Department of Mathematics and Statistics, Oakland University, Rochester, Michigan, USA
| | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
34
|
Della Torre S. Beyond the X Factor: Relevance of Sex Hormones in NAFLD Pathophysiology. Cells 2021; 10:2502. [PMID: 34572151 PMCID: PMC8470830 DOI: 10.3390/cells10092502] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue worldwide, being frequently associated with obesity, unbalanced dietary regimens, and reduced physical activity. Despite their greater adiposity and reduced physical activity, women show a lower risk of developing NAFLD in comparison to men, likely a consequence of a sex-specific regulation of liver metabolism. In the liver, sex differences in the uptake, synthesis, oxidation, deposition, and mobilization of lipids, as well as in the regulation of inflammation, are associated with differences in NAFLD prevalence and progression between men and women. Given the major role of sex hormones in driving hepatic sexual dimorphism, this review will focus on the role of sex hormones and their signaling in the regulation of hepatic metabolism and in the molecular mechanisms triggering NAFLD development and progression.
Collapse
Affiliation(s)
- Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
35
|
Pan R, Chen Y. Fat biology and metabolic balance: On the significance of sex. Mol Cell Endocrinol 2021; 533:111336. [PMID: 34090969 DOI: 10.1016/j.mce.2021.111336] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Obesity and its related metabolic disorders have become prevalent and fatal, which are faced by the entire human beings since decades. An energy equilibrium is urgently important for human metabolic health, which requires the participation of multiple organs, such as adipose tissues, liver and skeletal muscles. It seems that both sex and age play a role in the above processes. In this review, we focus on the sexual dimorphism in energy metabolism mediated by adipose tissues, including white and thermogenic (brown/beige) adipose tissues. Remarkably, past investigations have focused on targeting brown/beige adipose tissues to combat obesity because of their contributions to non-shivering thermogenesis. However, sex differences in the regulation of adipose tissue metabolism are likely overlooked. Particularly, increasing data show that males display more visceral fat than females, and females show increased visceral fat after menopause. Visceral adiposity is more deleterious and closely related to metabolic disorders, such as cardiovascular diseases. In this review, we discuss current findings on sexual dimorphism in WAT and BAT biology for a better metabolic balance in humans.
Collapse
Affiliation(s)
- Ruping Pan
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, PR China; Branch of National Clinical Research Center for Metabolic Diseases, Hubei, PR China.
| |
Collapse
|
36
|
Ibrahim A, Mahmoud UT, Ali MM, Ragab SMM. Evaluation of the subcapsular technique for primary closure castration in donkeys (Equus asinus). Sci Rep 2021; 11:14080. [PMID: 34234262 PMCID: PMC8263736 DOI: 10.1038/s41598-021-93585-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
This study described the subcapsular technique for primary closure castration in donkeys with special regard to its efficiency and welfare impacts. The study was conducted on twelve adult male donkeys, allocated randomly into two groups; subcapsular castration (SC) and open castration (OC) groups, whether the donkeys were subjected to surgical castration either by subcapsular or open castration techniques, respectively. Testosterone, cortisol, lactate, glucose, total cholesterol (TC), high density lipoprotein cholesterol (HDL-C), triglyceride (TG), and nitric oxide (NO) were measured before and after castration. Pain-associated behavioral activities were recorded post-castration. The SC was successfully performed in donkeys through a single paramedian scrotal incision. The SC was efficient as OC in reducing testosterone levels. The pain score decreased in the SC compared to the OC over time. The SC was an efficient and reliable technique for primary closure castration in donkeys with minimal postoperative complications and care and good cosmetic, physiological, and behavioral outcomes. It can be an alternative to other castration techniques in equines.
Collapse
Affiliation(s)
- Ahmed Ibrahim
- grid.252487.e0000 0000 8632 679XVeterinary Teaching Hospital, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526 Egypt
| | - Usama T. Mahmoud
- grid.252487.e0000 0000 8632 679XDepartment of Animal and Poultry Behavior and Management, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526 Egypt
| | - Magda M. Ali
- grid.252487.e0000 0000 8632 679XDepartment of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526 Egypt
| | - Sohair M. M. Ragab
- Department of Zoology, Faculty of Science, Assiut Uuniversity, Assiut, 71526 Egypt
| |
Collapse
|
37
|
Kim NR, David K, Corbeels K, Khalil R, Antonio L, Schollaert D, Deboel L, Ohlsson C, Gustafsson JÅ, Vangoitsenhoven R, Van der Schueren B, Decallonne B, Claessens F, Vanderschueren D, Dubois V. Testosterone Reduces Body Fat in Male Mice by Stimulation of Physical Activity Via Extrahypothalamic ERα Signaling. Endocrinology 2021; 162:bqab045. [PMID: 33674833 PMCID: PMC8140602 DOI: 10.1210/endocr/bqab045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Indexed: 12/21/2022]
Abstract
Testosterone (T) reduces male fat mass, but the underlying mechanisms remain elusive, limiting its clinical relevance in hypogonadism-associated obesity. Here, we subjected chemically castrated high-fat diet-induced adult obese male mice to supplementation with T or the nonaromatizable androgen dihydrotestosterone (DHT) for 20 weeks. Both hormones increased lean mass, thereby indirectly increasing oxygen consumption and energy expenditure. In addition, T but not DHT decreased fat mass and increased ambulatory activity, indicating a role for aromatization into estrogens. Investigation of the pattern of aromatase expression in various murine tissues revealed the absence of Cyp19a1 expression in adipose tissue while high levels were observed in brain and gonads. In obese hypogonadal male mice with extrahypothalamic neuronal estrogen receptor alpha deletion (N-ERαKO), T still increased lean mass but was unable to decrease fat mass. The stimulatory effect of T on ambulatory activity was also abolished in N-ERαKO males. In conclusion, our work demonstrates that the fat-burning action of T is dependent on aromatization into estrogens and is at least partially mediated by the stimulation of physical activity via extrahypothalamic ERα signaling. In contrast, the increase in lean mass upon T supplementation is mediated through the androgen receptor and indirectly leads to an increase in energy expenditure, which might also contribute to the fat-burning effects of T.
Collapse
Affiliation(s)
- Na Ri Kim
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Karel David
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Katrien Corbeels
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Rougin Khalil
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Leen Antonio
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Dieter Schollaert
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Ludo Deboel
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg 413 45, Sweden
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204-5056, USA
| | - Roman Vangoitsenhoven
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Bart Van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| | - Vanessa Dubois
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
38
|
Davidyan A, Pathak S, Baar K, Bodine SC. Maintenance of muscle mass in adult male mice is independent of testosterone. PLoS One 2021; 16:e0240278. [PMID: 33764986 PMCID: PMC7993603 DOI: 10.1371/journal.pone.0240278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/12/2021] [Indexed: 01/21/2023] Open
Abstract
Testosterone is considered a potent anabolic agent in skeletal muscle with a well-established role in adolescent growth and development in males. However, the role of testosterone in the regulation of skeletal muscle mass and function throughout the lifespan has yet to be fully established. While some studies suggest that testosterone is important for the maintenance of skeletal muscle mass, an understanding of the role this hormone plays in young, adult, and old males with normal and low serum testosterone levels is lacking. We investigated the role testosterone plays in the maintenance of muscle mass by examining the effect of orchiectomy-induced testosterone depletion in C57Bl6 male mice at ages ranging from early postnatal through old age (1.5-, 5-, 12-, and 24-month old mice). Following 28 days of testosterone depletion, we assessed mass and fiber cross-sectional-area (CSA) of the tibialis anterior, gastrocnemius, and quadriceps muscles. In addition, we measured global rates of protein synthesis and degradation using the SuNSET method, western blots, and enzyme activity assays. Twenty-eight days of testosterone depletion resulted in reduced muscle mass in the two youngest cohorts, but had no effect in the two oldest cohorts. Mean CSA decreased only in the youngest cohort and only in the tibialis anterior muscle. Testosterone depletion resulted in a general increase in proteasome activity at all ages. No change in protein synthesis was detected at the terminal time point. These data suggest that within physiological serum concentrations, testosterone may not be critical for the maintenance of muscle mass in mature male mice; however, in young mice testosterone is crucial for normal growth.
Collapse
Affiliation(s)
- Arik Davidyan
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, United States of America
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
| | - Suraj Pathak
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, United States of America
| | - Keith Baar
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, United States of America
- Department of Physiology and Membrane Biology University of California Davis, Davis, CA, United States of America
| | - Sue C. Bodine
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
39
|
Kim NR, Khalil R, David K, Antonio L, Schollaert D, Deboel L, Van Herck E, Wardenier N, Cools M, Decallonne B, Claessens F, Dubois V, Vanderschueren D. Novel model to study the physiological effects of temporary or prolonged sex steroid deficiency in male mice. Am J Physiol Endocrinol Metab 2021; 320:E415-E424. [PMID: 33308013 DOI: 10.1152/ajpendo.00401.2020] [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] [Indexed: 01/09/2023]
Abstract
Sex steroids are critical for skeletal development and maturation during puberty as well as for skeletal maintenance during adult life. However, the exact time during puberty when sex steroids have the highest impact as well as the ability of bone to recover from transient sex steroid deficiency is unclear. Surgical castration is a common technique to study sex steroid effects in rodents, but it is irreversible, invasive, and associated with metabolic and behavioral alterations. Here, we used a low dose (LD) or a high dose (HD) of gonadotropin-releasing hormone antagonist to either temporarily or persistently suppress sex steroid action in male mice, respectively. The LD group, a model for delayed puberty, did not show changes in linear growth or body composition, but displayed reduced trabecular bone volume during puberty, which fully caught up at adult age. In contrast, the HD group, representing complete pubertal suppression, showed a phenotype reminiscent of that observed in surgically castrated rodents. Indeed, HD animals exhibited severely impaired cortical and trabecular bone acquisition, decreased body weight and lean mass, and increased fat mass. In conclusion, we developed a rodent model of chemical castration that can be used as an alternative to surgical castration. Moreover, the transient nature of the intervention enables to study the effects of delayed puberty and reversibility of sex steroid deficiency.NEW & NOTEWORTHY We developed a rodent model of chemical castration, which can be used as an alternative to surgical castration. Moreover, the transient nature of the intervention enables to study the effects of delayed puberty and reversibility of sex steroid deficiency.
Collapse
Affiliation(s)
- Na Ri Kim
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Rougin Khalil
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Karel David
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Leen Antonio
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Dieter Schollaert
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Ludo Deboel
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Erik Van Herck
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Nele Wardenier
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Martine Cools
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Vanessa Dubois
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| |
Collapse
|
40
|
Sebo ZL, Rodeheffer MS. Testosterone metabolites differentially regulate obesogenesis and fat distribution. Mol Metab 2020; 44:101141. [PMID: 33307216 PMCID: PMC7772371 DOI: 10.1016/j.molmet.2020.101141] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/20/2020] [Accepted: 12/03/2020] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE Low testosterone in men (hypogonadism) is associated with obesity and type II diabetes. Testosterone replacement therapy has been shown to reverse these effects. However, the mechanisms by which testosterone regulates total fat mass, fat distribution, and metabolic health are unclear. In this study, we clarify the impact of hypogonadism on these parameters, as well as parse the role of testosterone from its downstream metabolites, dihydrotestosterone (DHT), and estradiol, in the regulation of depot-specific adipose tissue mass. METHODS To achieve this objective, we utilized mouse models of male hypogonadism coupled with hormone replacement therapy, magnetic resonance imaging (MRI), glucose tolerance tests, flow cytometry, and immunohistochemical techniques. RESULTS We observed that castrated mice develop increased fat mass, reduced muscle mass, and impaired glucose metabolism compared with gonadally intact males. Interestingly, obesity is further accelerated in castrated mice fed a high-fat diet, suggesting hypogonadism increases susceptibility to obesogenesis when dietary consumption of fat is elevated. By performing hormone replacement therapy in castrated mice, we show that testosterone impedes visceral and subcutaneous fat mass expansion. Testosterone-derived estradiol selectively blocks visceral fat growth, and DHT selectively blocks the growth of subcutaneous fat. These effects are mediated by depot-specific alterations in adipocyte size. We also show that high-fat diet-induced adipogenesis is elevated in castrated mice and that this can be rescued by androgen treatment. Obesogenic adipogenesis is also elevated in mice where androgen receptor activity is inhibited. CONCLUSIONS These data indicate that hypogonadism impairs glucose metabolism and increases obesogenic fat mass expansion through adipocyte hypertrophy and adipogenesis. In addition, our findings highlight distinct roles for testosterone, DHT, and estradiol in the regulation of total fat mass and fat distribution and reveal that androgen signaling blocks obesogenic adipogenesis in vivo.
Collapse
Affiliation(s)
- Zachary L Sebo
- Yale University, Department of Molecular, Cellular and Developmental Biology, USA
| | - Matthew S Rodeheffer
- Yale University, Department of Molecular, Cellular and Developmental Biology, USA; Department of Comparative Medicine, Yale University, USA; Department of Physiology, Yale University, USA; Yale Stem Cell Center, USA; Yale Program in Integrative Cell Signaling and Neurobiology of Metabolism, USA.
| |
Collapse
|
41
|
Hermoso DAM, Bizerra PFV, Constantin RP, Ishii-Iwamoto EL, Gilglioni EH. Association between metabolic syndrome, hepatic steatosis, and testosterone deficiency: evidences from studies with men and rodents. Aging Male 2020; 23:1296-1315. [PMID: 32406295 DOI: 10.1080/13685538.2020.1764927] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Testosterone is the predominant androgen in men and the lack of it can be a trigger to the development of the metabolic syndrome. Here we review the relationship between testosterone deficiency, metabolic syndrome, and hepatic steatosis reported by studies with men and rodents. The prevalence of metabolic syndrome and testosterone deficiency is higher among older subjects. Low total and free testosterone levels were positively associated with disturbs on energy metabolism, changes in body fat distribution, and body composition. Studies reported visceral fat accumulation in men with hypogonadism and castrated rats. Despite some contradictions, the association between higher adiposity, low testosterone, and metabolic syndrome was a common point among the studies. Few studies evaluated the hepatic steatosis and found an association with hypogonadism. Most of the studies with rodents combined the castration with a high-fat diet to study metabolic disturbs. The importance of proper levels of testosterone for energy metabolism homeostasis in men was also underlined by studies that investigated the metabolic effects of testosterone replacement therapy and androgen deprivation therapy.
Collapse
Affiliation(s)
- Danielle Aparecida Munhos Hermoso
- Departament of Biochemistry, Laboratory of Biological Oxidation and Laboratory of Experimental Steatosis, State University of Maringá, Maringá, Brazil
| | - Paulo Francisco Veiga Bizerra
- Departament of Biochemistry, Laboratory of Biological Oxidation and Laboratory of Experimental Steatosis, State University of Maringá, Maringá, Brazil
| | - Rodrigo Polimeni Constantin
- Departament of Biochemistry, Laboratory of Biological Oxidation and Laboratory of Experimental Steatosis, State University of Maringá, Maringá, Brazil
| | - Emy Luiza Ishii-Iwamoto
- Departament of Biochemistry, Laboratory of Biological Oxidation and Laboratory of Experimental Steatosis, State University of Maringá, Maringá, Brazil
| | - Eduardo Hideo Gilglioni
- Departament of Biochemistry, Laboratory of Biological Oxidation and Laboratory of Experimental Steatosis, State University of Maringá, Maringá, Brazil
| |
Collapse
|
42
|
Li J, Tang C, Zhao Q, Yang Y, Li F, Qin Y, Liu X, Yue X, Zhang J. Integrated lipidomics and targeted metabolomics analyses reveal changes in flavor precursors in psoas major muscle of castrated lambs. Food Chem 2020; 333:127451. [DOI: 10.1016/j.foodchem.2020.127451] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/06/2020] [Accepted: 06/27/2020] [Indexed: 02/07/2023]
|
43
|
Moderate walnut consumption improved lipid profile, steroid hormones and inflammation in trained elderly men: a pilot study with a randomized controlled trial. Biol Sport 2020; 38:245-252. [PMID: 34079169 PMCID: PMC8139345 DOI: 10.5114/biolsport.2020.97676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/27/2020] [Accepted: 07/22/2020] [Indexed: 11/17/2022] Open
Abstract
The present study aimed to investigate the effect of walnut consumption on lipid profile, steroid hormones and inflammation in trained elderly men performing concurrent (resistance and endurance) training. Twenty healthy elderly males were divided into two matched groups, in a randomized controlled trial, that trained three sessions per week: concurrent training + dietary walnut consumption (15 g/day for six weeks, CTW: n = 10); concurrent training + control diet (CT: n = 10). Fasting blood samples were taken 48 hours before and after intervention for biochemical assessments. levels of high-density lipoprotein (HDL) increased only in CTW compared to baseline (19.8%, p < 0.01). Total cholesterol (TC), low-density lipoprotein (LDL) and triglyceride (TG) levels significantly decreased only for CTW (i.e., 13%, 18%, and 18.5% at p < 0.01 for all). Testosterone (T) increased after the training compared to pre-training for CTW and CT (10.3%, p < 0.01, 4.27% p < 0.05, respectively), but the increase was significantly higher in CTW (p < 0.05). Serum cortisol (C) was lower for CTW compared to CT (p < 0.01). C-reactive protein (CRP) decreased in CTW in comparison with CT. The present study revealed that 6-week moderate walnut supplementation (15 g/day) improved lipid profile, steroid hormones and systematic inflammation in aged men performing concurrent training. These findings could be attributable to the potential effect of polyunsaturated fatty acids (PUFA) contained in walnut (linoleic acid, n-6; linolenic acid, n-3).
Collapse
|
44
|
Baik M, Jeong JY, Park SJ, Yoo SP, Lee JO, Lee JS, Haque MN, Lee HJ. Testosterone deficiency caused by castration increases adiposity in male rats in a tissue-specific and diet-dependent manner. GENES AND NUTRITION 2020; 15:14. [PMID: 32807074 PMCID: PMC7433145 DOI: 10.1186/s12263-020-00673-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 08/05/2020] [Indexed: 11/11/2022]
Abstract
Background Testosterone deficiency in men is clinically associated with the development of metabolic syndrome, which manifests as obesity, hepatic steatosis, and type-2 diabetes. We investigated the effects of castration-induced testosterone deficiency on body adiposity and the expression of genes related to lipid metabolism and glucose uptake and androgen signaling in male rats fed a normal diet (ND) or a high-fat diet (HFD). Methods Changes in lipid and glucose metabolism and androgen signaling were investigated at physiological and molecular levels in the muscle, liver, and adipose tissues of non-castrated and castrated rats under ND or HFD feeding. Results Castration-induced testosterone deficiency predisposed animals on ND to early development of fatty liver by activating fatty acid (FA) synthesis, whereas HFD activated hepatic FA uptake CD36 expression, leading to the development of hepatic steatosis. In rats fed ND, castration induced muscle fat accumulation by activating CD36 expression. In the subcutaneous fat of ND-fed rats, castration increased adiposity and the expression of FA synthesis-related genes, but it decreased glucose transporter gene expression. In the abdominal fat of rats fed ND, castration increased adiposity by upregulating FA synthesis-related genes, and HFD promoted adiposity by inducing FA uptake, glucose transporter, and FA synthesis-related gene expression. In rats fed ND, castration decreased body growth and muscle weight and downregulated the expression of genes androgen signaling in the longissimus dorsi muscle. Conclusions Testosterone deficiency increases adiposity in a tissue-specific and diet-dependent manner. Testosterone deficiency decreases body and muscle weights and downregulates androgen signaling.
Collapse
Affiliation(s)
- Myunggi Baik
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea. .,Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea. .,Institutes of Green Bio Science Technology, Pyeongchang-daero, Daehwa-myeon, Pyoengchang-gun, Gangwon-do, 25354, Republic of Korea.
| | - Jin Young Jeong
- National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| | - Seung Ju Park
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.,Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seon Pil Yoo
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.,Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jin Oh Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.,Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Sung Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.,Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Md Najmul Haque
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.,Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun-Jeong Lee
- National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, 55365, Republic of Korea
| |
Collapse
|
45
|
Cara AL, Myers MG, Elias CF. Lack of AR in LepRb Cells Disrupts Ambulatory Activity and Neuroendocrine Axes in a Sex-Specific Manner in Mice. Endocrinology 2020; 161:bqaa110. [PMID: 32609838 PMCID: PMC7383963 DOI: 10.1210/endocr/bqaa110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/26/2020] [Indexed: 11/19/2022]
Abstract
Disorders of androgen imbalance, such as hyperandrogenism in females or hypoandrogenism in males, increase risk of visceral adiposity, type 2 diabetes, and infertility. Androgens act upon androgen receptors (AR) which are expressed in many tissues. In the brain, AR are abundant in hypothalamic nuclei involved in regulation of reproduction and energy homeostasis, yet the role of androgens acting via AR in specific neuronal populations has not been fully elucidated. Leptin receptor (LepRb)-expressing neurons coexpress AR predominantly in hypothalamic arcuate and ventral premammillary nuclei (ARH and PMv, respectively), with low colocalization in other LepRb neuronal populations, and very low colocalization in the pituitary gland and gonads. Deletion of AR from LepRb-expressing cells (LepRbΔAR) has no effect on body weight, energy expenditure, and glucose homeostasis in male and female mice. However, LepRbΔAR female mice show increased body length later in life, whereas male LepRbΔAR mice show an increase in spontaneous ambulatory activity. LepRbΔAR mice display typical pubertal timing, estrous cycles, and fertility, but increased testosterone levels in males. Removal of sex steroid negative feedback action induced an exaggerated rise in luteinizing hormone in LepRbΔAR males and follicle-stimulating hormone in LepRbΔAR females. Our findings show that AR can directly affect a subset of ARH and PMv neurons in a sex-specific manner and demonstrate specific androgenic actions in the neuroendocrine hypothalamus.
Collapse
Affiliation(s)
- Alexandra L Cara
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Martin G Myers
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Department of Obstetrics and Gynaecology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
46
|
Liu Y, Horn JL, Banda K, Goodman AZ, Lim Y, Jana S, Arora S, Germanos AA, Wen L, Hardin WR, Yang YC, Coleman IM, Tharakan RG, Cai EY, Uo T, Pillai SPS, Corey E, Morrissey C, Chen Y, Carver BS, Plymate SR, Beronja S, Nelson PS, Hsieh AC. The androgen receptor regulates a druggable translational regulon in advanced prostate cancer. Sci Transl Med 2020; 11:11/503/eaaw4993. [PMID: 31366581 DOI: 10.1126/scitranslmed.aaw4993] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/23/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022]
Abstract
The androgen receptor (AR) is a driver of cellular differentiation and prostate cancer development. An extensive body of work has linked these normal and aberrant cellular processes to mRNA transcription; however, the extent to which AR regulates posttranscriptional gene regulation remains unknown. Here, we demonstrate that AR uses the translation machinery to shape the cellular proteome. We show that AR is a negative regulator of protein synthesis and identify an unexpected relationship between AR and the process of translation initiation in vivo. This is mediated through direct transcriptional control of the translation inhibitor 4EBP1. We demonstrate that lowering AR abundance increases the assembly of the eIF4F translation initiation complex, which drives enhanced tumor cell proliferation. Furthermore, we uncover a network of pro-proliferation mRNAs characterized by a guanine-rich cis-regulatory element that is particularly sensitive to eIF4F hyperactivity. Using both genetic and pharmacologic methods, we demonstrate that dissociation of the eIF4F complex reverses the proliferation program, resulting in decreased tumor growth and improved survival in preclinical models. Our findings reveal a druggable nexus that functionally links the processes of mRNA transcription and translation initiation in an emerging class of lethal AR-deficient prostate cancer.
Collapse
Affiliation(s)
- Yuzhen Liu
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jessie L Horn
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kalyan Banda
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Asha Z Goodman
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Yiting Lim
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sujata Jana
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sonali Arora
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Alexandre A Germanos
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Lexiaochuan Wen
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - William R Hardin
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Yu C Yang
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ilsa M Coleman
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Robin G Tharakan
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Elise Y Cai
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Takuma Uo
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98104, USA
| | - Smitha P S Pillai
- Comparative Medicine, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98915, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA 98915, USA
| | - Yu Chen
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Brett S Carver
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen R Plymate
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98104, USA
| | - Slobodan Beronja
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Departments of Medicine and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Andrew C Hsieh
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. .,Departments of Medicine and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
47
|
Corbeels K, Verlinden L, Lannoo M, Khalil R, Deleus E, Mertens A, Matthys C, Verstuyf A, Meulemans A, Vangoitsenhoven R, Carmeliet G, Van der Schueren B. The curious fate of bone following bariatric surgery: bone effects of sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB) in mice. Int J Obes (Lond) 2020; 44:2165-2176. [PMID: 32546862 DOI: 10.1038/s41366-020-0626-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/12/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Bone loss and increased fracture risk following bariatric surgery has been reported. We investigated whether the two most commonly performed surgeries, sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB), lead to bone loss. In addition, we examined whether fortification of the diet with calcium citrate prevents bone loss. METHODS We used mouse models for SG and RYGB and compared bone loss with a group of sham mice with similar weight loss. All groups were switched at the time of surgery to a low-fat diet (LFD). We also examined whether fortification of the diet with calcium citrate and vitamin D was able to prevent bone loss. RESULTS At 2 weeks we observed no major bone effects. However, at 8 weeks, both trabecular and cortical bone were lost to the same extent after SG and RYGB, despite increased calcium absorption and adequate serum levels of calcium, vitamin D, and parathyroid hormone (PTH). Diet fortification with calcium citrate and vitamin D was able to partially prevent bone loss. CONCLUSIONS Both SG and RYGB lead to excess bone loss, despite intestinal adaptations to increase calcium absorption. Fortifying the diet with calcium citrate and vitamin D partly prevented the observed bone loss. This finding emphasizes the importance of nutritional support strategies after bariatric surgery, but also affirms that the exact mechanisms leading to bone loss after bariatric surgery remain elusive and thus warrant further research.
Collapse
Affiliation(s)
- Katrien Corbeels
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.
| | - Lieve Verlinden
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Matthias Lannoo
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.,Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Rougin Khalil
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Ann Mertens
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.,Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Christophe Matthys
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.,Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Annemieke Verstuyf
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Ann Meulemans
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.,Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Roman Vangoitsenhoven
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.,Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Bart Van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.,Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
48
|
Kirlangic OF, Yilmaz-Oral D, Kaya-Sezginer E, Toktanis G, Tezgelen AS, Sen E, Khanam A, Oztekin CV, Gur S. The Effects of Androgens on Cardiometabolic Syndrome: Current Therapeutic Concepts. Sex Med 2020; 8:132-155. [PMID: 32201216 PMCID: PMC7261691 DOI: 10.1016/j.esxm.2020.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/31/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Cardiometabolic syndrome (CMS), as a bunch of metabolic disorders mainly characterized by type 2 diabetes mellitus (T2DM), hypertension, atherosclerosis, central adiposity, and abdominal obesity triggering androgen deficiency, is one of the most critical threats to men. Although many significant preclinical and clinical findings explain CMS, new approaches toward common pathophysiological mechanisms and reasonable therapeutic targets are lacking. AIM To gain a further understanding of the role of androgen levels in various facets of CMS such as the constellation of cardiometabolic risk factors including central adiposity, dyslipidemia, insulin resistance, diabetes, and arterial hypertension and to define future directions for development of effective therapeutic modalities. METHODS Clinical and experimental data were searched through scientific literature databases (PubMed) from 2009 to October 2019. MAIN OUTCOME MEASURE Evidence from basic and clinical research was gathered with regard to the causal impact and therapeutic roles of androgens on CMS. RESULTS There are important mechanisms implicated in androgen levels and the risk of CMS. Low testosterone levels have many signs and symptoms on cardiometabolic and glycometabolic risks as well as abdominal obesity in men. CLINICAL IMPLICATIONS The implications of the findings can shed light on future improvements in androgen levels and add potentially predictive risk for CMS, as well as T2DM, abdominal obesity to guide clinical management in the early stage. STRENGTHS & LIMITATIONS This comprehensive review refers to the association between androgens and cardiovascular health. A limitation of this study is the lack of large, prospective population-based studies that analyze the effects of testosterone treatment on CMS or mortality. CONCLUSION Low testosterone levels have several common features with metabolic syndrome. Thus, testosterone may have preventive role in the progress of metabolic syndrome and subsequent T2DM, abdominal obesity, and cardiovascular disease and likely affect aging men's health mainly through endocrine and vascular mechanisms. Further studies are necessary to evaluate the therapeutic interventions directed at preventing CMS in men. Kirlangic OF, Yilmaz-Oral D, Kaya-Sezginer E, et al. The Effects of Androgens on Cardiometabolic Syndrome: Current Therapeutic Concepts. Sex Med 2020;8:132-155.
Collapse
Affiliation(s)
- Omer Faruk Kirlangic
- University of Health Sciences, Gulhane Faculty of Medicine, Department of Medical Biochemistry, Ankara, Turkey
| | - Didem Yilmaz-Oral
- Cukurova University, Faculty of Pharmacy, Department of Pharmacology, Adana, Turkey
| | - Ecem Kaya-Sezginer
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| | - Gamze Toktanis
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | | | - Ekrem Sen
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | - Armagan Khanam
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | - Cetin Volkan Oztekin
- Department of Urology, Faculty of Medicine, University of Kyrenia, Girne, Turkish Republic of North Cyprus, Mersin 10, Turkey
| | - Serap Gur
- Ankara University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey.
| |
Collapse
|
49
|
Harada N, Hanada K, Minami Y, Kitakaze T, Ogata Y, Tokumoto H, Sato T, Kato S, Inui H, Yamaji R. Role of gut microbiota in sex- and diet-dependent metabolic disorders that lead to early mortality of androgen receptor-deficient male mice. Am J Physiol Endocrinol Metab 2020; 318:E525-E537. [PMID: 32017595 DOI: 10.1152/ajpendo.00461.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gut microbiota is involved in metabolic disorders induced by androgen deficiency after sexual maturation in males (late-onset hypogonadism). However, its role in the energy metabolism of congenital androgen deficiency (e.g., androgen-insensitive syndrome) remains elusive. Here, we examined the link between the gut microbiota and metabolic disease symptoms in androgen receptor knockout (ARKO) mouse by administering high-fat diet (HFD) and/or antibiotics. HFD-fed male, but not standard diet-fed male or HFD-fed female, ARKO mice exhibited increased feed efficiency, obesity with increased visceral adipocyte mass and hypertrophy, hepatic steatosis, glucose intolerance, insulin resistance, and loss of thigh muscle. In contrast, subcutaneous fat mass accumulated in ARKO mice irrespective of the diet and sex. Notably, all HFD-dependent metabolic disorders observed in ARKO males were abolished after antibiotics administration. The ratios of fecal weight-to-food weight and cecum weight-to-body weight were specifically reduced by ARKO in HFD-fed males. 16S rRNA sequencing of fecal microbiota from HFD-fed male mice revealed differences in microbiota composition between control and ARKO mice. Several genera or species (e.g., Turicibacter and Lactobacillus reuteri, respectively) were enriched in ARKO mice, and antibiotics treatment spoiled the changes. Furthermore, the life span of HFD-fed ARKO males was shorter than that of control mice, indicating that androgen deficiency causes metabolic dysfunctions leading to early death. These findings also suggest that AR signaling plays a role in the prevention of metabolic dysfunctions, presumably by influencing the gut microbiome, and improve our understanding of health consequences in subjects with hypogonadism and androgen insensitivity.
Collapse
Affiliation(s)
- Naoki Harada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Kazuki Hanada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Yukari Minami
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Tomoya Kitakaze
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Yoshiyuki Ogata
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Hayato Tokumoto
- Division of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Takashi Sato
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Shigeaki Kato
- Graduate School of Science and Engineering, Iryo Sosei University, Iwaki, Fukushima, Japan
| | - Hiroshi Inui
- Department of Nutrition, College of Health and Human Sciences, Osaka Prefecture University, Habikino, Osaka, Japan
| | - Ryoichi Yamaji
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| |
Collapse
|
50
|
Crucial role of androgen receptor in resistance and endurance trainings-induced muscle hypertrophy through IGF-1/IGF-1R- PI3K/Akt- mTOR pathway. Nutr Metab (Lond) 2020; 17:26. [PMID: 32256674 PMCID: PMC7106900 DOI: 10.1186/s12986-020-00446-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background Androgen receptor (AR) has been reported to play vital roles in exercise-induced increase of muscle mass in rats, but needs to be further verified and the mechanism behind remains unclear. As AR target genes, insulin growth factor-1 (IGF-1) and IGF-1 receptor (IGF-1R) promote muscle hypertrophy through activating PI3K/Akt- mammalian target of rapamycin (mTOR) pathway, a classic pathway of muscle hypertrophy. So the main purpose of this study was using AR antagonist flutamide to demonstrate AR’s effect on training-induced muscle hypertrophy and its possible mechanism: IGF-1/IGF-1R- PI3K/Akt- mTOR pathway? Methods Forty-eight Sprague Dawley male rats aged 7 weeks were randomly divided into six groups: control (C), flutamide (F), resistance training (R), resistance training plus flutamide (R + F), endurance training (E), and endurance training plus flutamide (E + F) groups. Flutamide was used to block AR in rats. Rats in R and R + F groups fulfilled 3 weeks of ladder climbing with progressively increased load, while E and E + F rats completed 3-week moderate intensity aerobic exercise on a treadmill. The relative muscle mass (muscle mass/body weight) of rats was detected. Serum levels of testosterone and IGF-1 of rats were determined by ELISA, and mRNA levels of IGF-1R and mTOR in muscles by real-time PCR. Protein levels of AR, IGF-1, IGF-1R, mTOR, PI3K, Akt, p-PI3K and p-Akt in muscles were detected by Western blot. Results (1) The training-induced rise in the relative muscle mass and the expression levels of AR were only found in the gastrocnemius of R rats and in the soleus of E rats (selective muscle hypertrophy), which were blocked by flutamide. (2) Serum testosterone in the R and E rat were increased, and flutamide exerted no effect. (3) The levels of IGF-1, IGF-1R and mTOR as well as the activities of PI3K and Akt were enhanced selectively (in the gastrocnemius of R rats and in the soleus of E rats), which were reduced by flutamide. Conclusions: AR exerted an essential role in both resistance training and endurance training-induced muscle hypertrophy, which was mediated at least partly through IGF-1/IGF-1R- PI3K/Akt- mTOR pathway.
Collapse
|