1
|
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
|
2
|
Poklukar K, Erbežnik A, Fazarinc G, Kress K, Batorek-Lukač N, Škrlep M, Stefanski V, Čandek-Potokar M, Vrecl M. Effect of castration method on porcine skeletal muscle fiber traits and transcriptome profiles. Vet Anim Sci 2024; 25:100383. [PMID: 39184227 PMCID: PMC11342880 DOI: 10.1016/j.vas.2024.100383] [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] [Indexed: 08/27/2024] Open
Abstract
This study examined the effects of immunocastration and surgical castration on the histomorphometric and transcriptome traits of the porcine skeletal muscle. We hypothesized that the differences in duration of androgen deprivation resulting from different castration methods influence skeletal muscle biology in a muscle-specific manner. This was tested by analyzing samples of m. longissimus dorsi (LD) and m. semispinalis capitis (SSC) from immunocastrated (IC; n = 12), entire male (EM; n = 12), and surgically castrated (SC; n = 12) pigs using enzyme/immunohistochemical classification and histomorphometric analysis of myofibers, quantitative PCR, and RNA sequencing. The results confirmed the distinctive histomorphometric profiles of LD and SSC and the castration method related muscle-specific effects at the histomorphometric and transcriptome levels. Long-term androgen deficiency (surgical castration) significantly reduced the proportion of fast-twitch type IIa myofibers in LD (P < 0.05), whereas short-term androgen deprivation (immunocastration) reduced the cross-sectional area of oxidative type I myofibers in SSC (P < 0.05). At the transcriptional level, glycolytic LD adapted to long- and short-term androgen deprivation by upregulating genes controlling myoblast proliferation and differentiation to maintain fiber size. In contrast, increased protein degradation through the ubiquitin ligase-mediated atrophy pathway (significantly increased TRIM63 and FBXO32 expression; P < 0.05) could underly reduced cross-sectional area of type I myofibers in the oxidative SSC in IC. Potential candidate genes (HK2, ARID5B, SERPINE1, and SCD) linked to specific metabolic profiles and meat quality traits were also identified in IC, providing a foundation for studying the effects of immunocastration on skeletal muscle fiber and carcass/meat quality traits.
Collapse
Affiliation(s)
- Klavdija Poklukar
- Agricultural Institute of Slovenia (KIS), Hacquetova ulica 17, SI-1000, Ljubljana, Slovenia
| | - Anja Erbežnik
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva ulica 60, SI-1000, Ljubljana, Slovenia
| | - Gregor Fazarinc
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva ulica 60, SI-1000, Ljubljana, Slovenia
| | - Kevin Kress
- University of Hohenheim, Garbenstraße 17, 70599, Stuttgart, Germany
| | - Nina Batorek-Lukač
- Agricultural Institute of Slovenia (KIS), Hacquetova ulica 17, SI-1000, Ljubljana, Slovenia
| | - Martin Škrlep
- Agricultural Institute of Slovenia (KIS), Hacquetova ulica 17, SI-1000, Ljubljana, Slovenia
| | - Volker Stefanski
- University of Hohenheim, Garbenstraße 17, 70599, Stuttgart, Germany
| | - Marjeta Čandek-Potokar
- Agricultural Institute of Slovenia (KIS), Hacquetova ulica 17, SI-1000, Ljubljana, Slovenia
- Faculty of Agriculture and Life Sciences, University of Maribor, Pivola 10, SI-2311, Hoče, Slovenia
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva ulica 60, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
3
|
Sakai H, Imai Y. Cell-specific functions of androgen receptor in skeletal muscles. Endocr J 2024; 71:437-445. [PMID: 38281756 DOI: 10.1507/endocrj.ej23-0691] [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] [Indexed: 01/30/2024] Open
Abstract
Androgens play a vital role not only in promoting the development of male sexual characteristics but also in exerting diverse physiological effects, including the regulation of skeletal muscle growth and function. Given that the effects of androgens are mediated through androgen receptor (AR) binding, an understanding of AR functionality is crucial for comprehending the mechanisms of androgen action on skeletal muscles. Drawing from insights gained using conditional knockout mouse models facilitated by Cre/loxP technology, we review the cell-specific functions of AR in skeletal muscles. We focus on three specific cell populations expressing AR within skeletal muscles: skeletal muscle cells, responsible for muscle contraction; satellite cells, which are essential stem cells contributing to the growth and regeneration of skeletal muscles; and mesenchymal progenitors, situated in interstitial areas and playing a crucial role in muscle homeostasis. Furthermore, the indirect effects of androgens on skeletal muscle through extra-muscle tissue are essential, especially for the regulation of skeletal muscle mass. The regulation of genes by AR varies across different cell types and contexts, including homeostasis, regeneration and hypertrophy of skeletal muscles. The varied mechanisms orchestrated by AR collectively influence the physiology of skeletal muscles.
Collapse
Affiliation(s)
- Hiroshi Sakai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Ehime 791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Ehime 791-0295, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Ehime 791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Ehime 791-0295, Japan
| |
Collapse
|
4
|
Takahashi F, Baba T, Christianto A, Yanai S, Lee-Okada HC, Ishiwata K, Nakabayashi K, Hata K, Ishii T, Hasegawa T, Yokomizo T, Choi MH, Morohashi KI. Development of sexual dimorphism of skeletal muscles through the adrenal cortex, caused by androgen-induced global gene suppression. Cell Rep 2024; 43:113715. [PMID: 38306273 DOI: 10.1016/j.celrep.2024.113715] [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: 04/12/2023] [Revised: 12/08/2023] [Accepted: 01/11/2024] [Indexed: 02/04/2024] Open
Abstract
The zona fasciculata (zF) in the adrenal cortex contributes to multiple physiological actions through glucocorticoid synthesis. The size, proliferation, and glucocorticoid synthesis characteristics are all female biased, and sexual dimorphism is established by androgen. In this study, transcriptomes were obtained to unveil the sex differentiation mechanism. Interestingly, both the amount of mRNA and the expressions of nearly all genes were higher in females. The expression of Nr5a1, which is essential for steroidogenic cell differentiation, was also female biased. Whole-genome studies demonstrated that NR5A1 regulates nearly all gene expression directly or indirectly. This suggests that androgen-induced global gene suppression is potentially mediated by NR5A1. Using Nr5a1 heterozygous mice, whose adrenal cortex is smaller than the wild type, we demonstrated that the size of skeletal muscles is possibly regulated by glucocorticoid synthesized by zF. Taken together, considering the ubiquitous presence of glucocorticoid receptors, our findings provide a pathway for sex differentiation through glucocorticoid synthesis.
Collapse
Affiliation(s)
- Fumiya Takahashi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Baba
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Antonius Christianto
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shogo Yanai
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hyeon-Cheol Lee-Okada
- Department of Biochemistry, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Keisuke Ishiwata
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-0074, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-0074, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, Research Institute, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-0074, Japan; Department of Human Molecular Genetics, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tomohiro Ishii
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Man Ho Choi
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Ken-Ichirou Morohashi
- Department of Systems Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Internal Medicine, Kurume University School of Medicine, 67 Asahimachi, Kurume 830-0011, Japan.
| |
Collapse
|
5
|
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
|
6
|
Hosoi T, Yakabe M, Hashimoto S, Akishita M, Ogawa S. The roles of sex hormones in the pathophysiology of age-related sarcopenia and frailty. Reprod Med Biol 2024; 23:e12569. [PMID: 38476959 PMCID: PMC10927916 DOI: 10.1002/rmb2.12569] [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: 12/30/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Background Sarcopenia is an age-related condition characterized by a progressive and systemic decline in skeletal muscle mass, quality, and strength. The incidence of sarcopenia contains sex-specific aspects, indicating the contribution of sex hormones to its pathophysiology. This review focuses on changing trends in sarcopenia, discusses alterations in definitions and diagnostic criteria, and emphasizes the association between sarcopenia and sex hormones. Methods A literature search was performed on PubMed for related articles published between 1997 and December 2023 using appropriate keywords. Main Findings Results Advances in research have emphasized the significance of muscle quality and strength over muscle mass, resulting in new diagnostic criteria for sarcopenia. Androgens demonstrated anabolic effects on skeletal muscles and played a significant role in the pathophysiology of sarcopenia. In clinical settings, androgen replacement therapy has exhibited certain positive outcomes for treating sarcopenia, despite concerns about potential side effects. Conversely, estrogen is involved in skeletal muscle maintenance, but the detailed mechanisms remain unclear. Moreover, results regarding the clinical application of estrogen replacement therapy for treating sarcopenia remained inconsistent. Conclusion The elucidation of molecular mechanisms that involve sex hormones is eagerly awaited for novel therapeutic interventions for sarcopenia.
Collapse
Affiliation(s)
- Tatsuya Hosoi
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Mitsutaka Yakabe
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Seiji Hashimoto
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Sumito Ogawa
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| |
Collapse
|
7
|
Knapp M, Supruniuk E, Górski J. Myostatin and the Heart. Biomolecules 2023; 13:1777. [PMID: 38136649 PMCID: PMC10741510 DOI: 10.3390/biom13121777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Myostatin (growth differentiation factor 8) is a member of the transforming growth factor-β superfamily. It is secreted mostly by skeletal muscles, although small amounts of myostatin are produced by the myocardium and the adipose tissue as well. Myostatin binds to activin IIB membrane receptors to activate the downstream intracellular canonical Smad2/Smad3 pathway, and additionally acts on non-Smad (non-canonical) pathways. Studies on transgenic animals have shown that overexpression of myostatin reduces the heart mass, whereas removal of myostatin has an opposite effect. In this review, we summarize the potential diagnostic and prognostic value of this protein in heart-related conditions. First, in myostatin-null mice the left ventricular internal diameters along with the diastolic and systolic volumes are larger than the respective values in wild-type mice. Myostatin is potentially secreted as part of a negative feedback loop that reduces the effects of the release of growth-promoting factors and energy reprogramming in response to hypertrophic stimuli. On the other hand, both human and animal data indicate that myostatin is involved in the development of the cardiac cachexia and heart fibrosis in the course of chronic heart failure. The understanding of the role of myostatin in such conditions might initiate a development of targeted therapies based on myostatin signaling inhibition.
Collapse
Affiliation(s)
- Małgorzata Knapp
- Department of Cardiology, Medical University of Białystok, 15-276 Białystok, Poland
| | - Elżbieta Supruniuk
- Department of Physiology, Medical University of Białystok, 15-222 Białystok, Poland;
| | - Jan Górski
- Department of Health Sciences, University of Łomża, 18-400 Łomża, Poland;
| |
Collapse
|
8
|
Kumar A, Prajapati P, Raj V, Kim SC, Mishra V, Raorane CJ, Raj R, Kumar D, Kushwaha S. Salbutamol ameliorates skeletal muscle wasting and inflammatory markers in streptozotocin (STZ)-induced diabetic rats. Int Immunopharmacol 2023; 124:110883. [PMID: 37666067 DOI: 10.1016/j.intimp.2023.110883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/13/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
Diabetes accelerates muscle atrophy, leading to the deterioration of skeletal muscles. This study aimed to assess the potential of the β2-adrenoceptor agonist, salbutamol (SLB), to alleviate muscle atrophy in streptozotocin (STZ)-induced diabetic rats. Male Sprague Dawley rats were randomized into four groups (n=6): control, SLB, STZ (55 mg/kg, single i.p.), and STZ + SLB (6 mg/kg, orally for 4 weeks). After the final SLB dose, animals underwent tests to evaluate muscle strength and coordination, including forelimb grip strength, wire-hanging, actophotometer, rotarod, and footprint assessments. Rats were then sacrificed, and serum and gastrocnemius (GN) muscles were collected for further analysis. Serum evaluations included proinflammatory markers (tumor necrosis factor α, interleukin-1β, interleukin-6), muscle markers (creatine kinase, myostatin), testosterone, and lipidemic markers. Muscle oxidative stress (malonaldehyde, protein carbonyl), antioxidants (glutathione, catalase, superoxide dismutase), and histology were also performed. Additionally, 1H nuclear magnetic resonance serum profiling was conducted. SLB notably enhanced muscle grip strength, coordination, and antioxidant levels, while reduced proinflammatory markers and oxidative stress in STZ-induced diabetic rats. Reduced serum muscle biomarkers, increased testosterone, restored lipidemic levels, and improved muscle cellular architecture indicated SLB's positive effect on muscle condition in diabetic rats. Metabolomics profiling revealed that the STZ group significantly increased the phenylalanine-to-tyrosine ratio (PTR), lactate-to-pyruvate ratio (LPR), acetate, succinate, isobutyrate, and histidine. SLB administration restored these perturbed serum metabolites in the STZ-induced diabetic group. In conclusion, salbutamol significantly protected against skeletal muscle wasting in STZ-induced diabetic rats.
Collapse
Affiliation(s)
- Anand Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Priyanka Prajapati
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Vinit Raj
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Seong-Cheol Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India.
| | | | - Ritu Raj
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India
| | - Dinesh Kumar
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow 226014, India
| | - Sapana Kushwaha
- National Institutes of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow 226002, India.
| |
Collapse
|
9
|
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: 5] [Impact Index Per Article: 2.5] [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
|
10
|
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: 1] [Impact Index Per Article: 0.5] [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
|
11
|
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
|
12
|
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
|
13
|
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: 60] [Impact Index Per Article: 30.0] [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
|
14
|
Hahn AW, Siddiqui BA, Leo J, Dondossola E, Basham KJ, Miranti CK, Frigo DE. Cancer Cell-Extrinsic Roles for the Androgen Receptor in Prostate Cancer. Endocrinology 2023; 164:bqad078. [PMID: 37192413 PMCID: PMC10413433 DOI: 10.1210/endocr/bqad078] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023]
Abstract
Given the central role of the androgen receptor (AR) in prostate cancer cell biology, AR-targeted therapies have been the backbone of prostate cancer treatment for over 50 years. New data indicate that AR is expressed in additional cell types within the tumor microenvironment. Moreover, targeting AR for the treatment of prostate cancer has established side effects such as bone complications and an increased risk of developing cardiometabolic disease, indicating broader roles for AR. With the advent of novel technologies, such as single-cell approaches and advances in preclinical modeling, AR has been identified to have clinically significant functions in other cell types. In this mini-review, we describe new cancer cell-extrinsic roles for AR within the tumor microenvironment as well as systemic effects that collectively impact prostate cancer progression and patient outcomes.
Collapse
Affiliation(s)
- Andrew W Hahn
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bilal A Siddiqui
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Javier Leo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Cindy K Miranti
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| | - Daniel E Frigo
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
15
|
Huang R, Kibschull M, Briollais L, Pausova Z, Murphy K, Kingdom J, Lye S, Luo ZC. Cord blood myostatin concentrations by gestational diabetes mellitus and fetal sex. Front Endocrinol (Lausanne) 2023; 14:1018779. [PMID: 36875483 PMCID: PMC9975152 DOI: 10.3389/fendo.2023.1018779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 01/25/2023] [Indexed: 02/17/2023] Open
Abstract
INTRODUCTION Myostatin is a member of the transforming growth factor β superfamily, and is mainly secreted from skeletal muscle. Animal studies have demonstrated that deficiency in myostatin promotes muscle growth and protects against insulin resistance. In humans, gestational diabetes mellitus (GDM) affects fetal insulin sensitivity. Females are more insulin resistant and weigh less than males at birth. We sought to assess whether cord blood myostatin concentrations vary by GDM and fetal sex, and the associations with fetal growth factors. METHODS In a study of 44 GDM and 66 euglycemic mother-newborn dyads, myostatin, insulin, proinsulin, insulin-like growth factor (IGF)-1, IGF-2 and testosterone were measured in cord blood samples. RESULTS Cord blood myostatin concentrations were similar in GDM vs. euglycemic pregnancies (mean ± SD: 5.5 ± 1.4 vs. 5.8 ± 1.4 ng/mL, P=0.28), and were higher in males vs. females (6.1 ± 1.6 vs. 5.3 ± 1.0 ng/mL, P=0.006). Adjusting for gestational age, myostatin was negatively correlated with IGF-2 (r=-0.23, P=0.02), but not correlated with IGF-1 (P=0.60) or birth weight (P=0.23). Myostatin was strongly correlated with testosterone in males (r=0.56, P<0.001), but not in females (r=-0.08, P=0.58) (test for difference in r, P<0.001). Testosterone concentrations were higher in males vs. females (9.5 ± 6.4 vs. 7.1 ± 4.0 nmol/L, P=0.017), and could explain 30.0% (P=0.039) of sex differences in myostatin concentrations. DISCUSSION The study is the first to demonstrate that GDM does not impact cord blood myostatin concentration, but fetal sex does. The higher myostatin concentrations in males appear to be partly mediated by higher testosterone concentrations. These findings shed novel insight on developmental sex differences in insulin sensitivity regulation relevant molecules.
Collapse
Affiliation(s)
- Rong Huang
- Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Mount Sinai Hospital, and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mark Kibschull
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Laurent Briollais
- Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Mount Sinai Hospital, and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Zdenka Pausova
- The Hospital for Sick Children, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Kellie Murphy
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - John Kingdom
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Stephen Lye
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Zhong-Cheng Luo
- Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Mount Sinai Hospital, and Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- *Correspondence: Zhong-Cheng Luo,
| |
Collapse
|
16
|
Castelli F, Valero-Breton M, Hernandez M, Guarda F, Cornejo J, Cabello-Verrugio C, Cabrera D. Regulatory Mechanisms of Muscle Mass: The Critical Role of Resistance Training in Children and Adolescent. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:21-34. [PMID: 36280657 DOI: 10.1007/5584_2022_743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Muscle mass and strength are subjected to several regulations. We found endocrine signals such as growth hormone, insulin-like growth factor 1, testosterone, thyroid hormones, and glucocorticoids among them. Neural inputs also influence muscle development, modulating mass and strength. Among the external stimuli that modulate these muscular features is physical training such as resistance and endurance training. Specifically, resistance training can mediate an increase in muscle mass by hypertrophy in adults, but the effects in children and adolescents are full of myths for most of the population. However, the evidence shows that the impact of resistance training on children and adolescents is clear and provides a wide range of benefits. However, qualified professionals must be available since exercise prescription and subsequent supervision must follow this population's abilities, needs, and interests.
Collapse
Affiliation(s)
- F Castelli
- Experimental Hepatology Laboratory, Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Valero-Breton
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - M Hernandez
- Experimental Hepatology Laboratory, Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F Guarda
- Departmento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J Cornejo
- Experimental Hepatology Laboratory, Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile.
| | - D Cabrera
- Experimental Hepatology Laboratory, Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
- School of Medicine, Faculty of Medical Sciences, Universidad Bernardo O Higgins, Santiago, Chile.
| |
Collapse
|
17
|
高山 賢. [Recent advances in the sex steroid hormone action involved in the development of dementia and frailty]. Nihon Ronen Igakkai Zasshi 2022; 59:430-445. [PMID: 36476689 DOI: 10.3143/geriatrics.59.430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- 賢一 高山
- 東京都健康長寿医療センター研究所老化機構研究チームシステム加齢医学
| |
Collapse
|
18
|
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
|
19
|
El Kharraz S, Dubois V, Launonen KM, Helminen L, Palvimo JJ, Libert C, Smeets E, Moris L, Eerlings R, Vanderschueren D, Helsen C, Claessens F. N/C Interactions Are Dispensable for Normal In Vivo Functioning of the Androgen Receptor in Male Mice. Endocrinology 2022; 163:6652495. [PMID: 35908178 PMCID: PMC9756762 DOI: 10.1210/endocr/bqac104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Indexed: 11/19/2022]
Abstract
The androgen receptor (AR) plays a central role in the development and maintenance of the male phenotype. The binding of androgens to the receptor induces interactions between the carboxyterminal ligand-binding domain and the highly conserved 23FQNLF27 motif in the aminoterminal domain. The role of these so-called N/C interactions in AR functioning is debated. In vitro assays show that mutating the AR in the 23FQNLF27 motif (called ARNoC) attenuates the AR transactivation of reporter genes, has no effect on ligand binding, but does affect protein-protein interactions with several AR coregulators. To test the in vivo relevance of the N/C interaction, we analyzed the consequences of the genomic introduction of the ARNoC mutation in mice. Surprisingly, the ARNoC/Y mice show a normal male development, with unaffected male anogenital distance and normal accessory sex glands, male circulating androgen levels, body composition, and fertility. The responsiveness of androgen target genes in kidney, prostate, and testes was also unaffected. We thus conclude that the N/C interactions in the AR are not essential for the development of a male phenotype under normal physiological conditions.
Collapse
Affiliation(s)
- Sarah El Kharraz
- Correspondence: Frank Claessens, PhD, Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium. . Reprint requests can be sent to or
| | - Vanessa Dubois
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, 3000, Belgium
- Department of Basic and Applied Medical Sciences, Basic and Translational Endocrinology, Ghent University, Ghent, 9000, Belgium
| | - Kaisa-Mari Launonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Laura Helminen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, 70210, Finland
| | - Claude Libert
- VIB Center for Inflammation Research, VIB, Ghent, 9052, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Ghent, 9052, Belgium
| | - Elien Smeets
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Lisa Moris
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Roy Eerlings
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Dirk Vanderschueren
- Department of Chronic Diseases and Metabolism, Clinical and Experimental Endocrinology, KU Leuven, Leuven, 3000, Belgium
| | - Christine Helsen
- Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium
| | - Frank Claessens
- Correspondence: Frank Claessens, PhD, Department of Cellular and Molecular Medicine, Molecular Endocrinology Laboratory, KU Leuven, Leuven, 3000, Belgium. . Reprint requests can be sent to or
| |
Collapse
|
20
|
Ahmad I, Newell-Fugate AE. Androgen and androgen receptor control of mitochondrial function. Am J Physiol Cell Physiol 2022; 323:C835-C846. [PMID: 35704694 DOI: 10.1152/ajpcell.00205.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effects of androgens have been extensively studied in a variety of organs and cell types with increasing focus on the sexually dimorphic role androgens play not only with respect to cellular functions but also in metabolism. Although the classical mechanism of androgen action is via ligand-dependent binding with the nuclear transcription factor, androgen receptor (AR), cytosolic AR can also activate second messenger signaling pathways. Given that cytosolic AR can signal in this manner, there has been increased interest in the mechanisms by which androgens may control cellular organelle function. This review highlights the effects that androgens have on mitochondrial structure and function with emphasis on biogenesis, fusion/fission, mitophagy, bioenergetics (oxidative phosphorylation), and reactive oxygen species production. There are a number of publications on the effects of androgens in these general areas of mitochondrial function. However, the precise mechanisms by which androgens cause these effects are not known. Additionally, given that the nucleus and mitochondria work in tandem to control mitochondrial function and the mitochondria has its own DNA, future research efforts should focus on the direct, mechanistic effects of androgens on mitochondrial function.
Collapse
Affiliation(s)
- Irshad Ahmad
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Annie E Newell-Fugate
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
21
|
Arrieta H, Rezola-Pardo C, Sanz B, Virgala J, Lacunza-Zumeta M, Rodriguez-Larrad A, Irazusta J. Improving the Identification of Frailty in Long-Term Care Residents: A Cross-Sectional Study. Biol Res Nurs 2022; 24:530-540. [PMID: 35574636 DOI: 10.1177/10998004221100797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE To compare the capacity of blood myostatin concentration and physical, cognitive, and affective function tests to predict frailty among long-term care (LTC) residents. METHODS This cross-sectional analysis used baseline data from three randomized controlled trials involving 260 older adults in 14 LTC centers. Serum myostatin levels were analyzed by enzyme-linked immunosorbent assay. Frailty, physical fitness, cognitive and affective functions were assessed using validated tests and scales. RESULTS The Timed Up and Go, gait speed, 6-minute walk, and Berg Balance Scale had excellent capabilities in identifying frail individuals in accordance with Fried's Frailty Phenotype (FFP). The best tests for identifying frailty in accordance with the Clinical Frailty Scale (CFS) were Timed Up and Go and Berg Balance Scale. For the Tilburg Frailty Indicator (TFI), the best tests were Quality of Life in Alzheimer's Disease (QoL-AD) and Goldberg Anxiety. Myostatin, along with physical, cognitive, and affective function tests, improved the capability of the hand grip, arm-curl, Montreal Cognitive Assessment, Goldberg Anxiety, Goldberg Depression, and QoL-AD to identify frailty according to FFP, while myostatin improved CFS-defined frailty identification by the hand grip, arm-curl, 6-minute walk test, Berg Balance Scale, 30-second chair-stand, gait speed, Montreal Cognitive Assessment, Goldberg Anxiety, and De Jong-Gierveld Loneliness Scale. CONCLUSION Among LTC residents, serum myostatin was associated with being frail according to FFP and CFS. However, this measure was less discriminating of frailty than physical fitness tests (for FFP and CFS) and affective function parameters (for TFI). However, evaluated concurrently with physical, cognitive, and affective parameters, myostatin improved the capabilities of these measures to predict CFS-defined frailty.
Collapse
Affiliation(s)
- Haritz Arrieta
- Department of Nursing II, Faculty of Medicine and Nursing, 83067University of the Basque Country (UPV/EHU), Donostia-San Sebastián, Gipuzkoa, Spain
| | - Chloe Rezola-Pardo
- Department of Physiology, Faculty of Medicine and Nursing, 83067University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Begoña Sanz
- Department of Physiology, Faculty of Medicine and Nursing, 83067University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Janire Virgala
- Department of Physiology, Faculty of Medicine and Nursing, 83067University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Ana Rodriguez-Larrad
- Department of Physiology, Faculty of Medicine and Nursing, 83067University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jon Irazusta
- Department of Physiology, Faculty of Medicine and Nursing, 83067University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
22
|
Sanz B, Rezola-Pardo C, Arrieta H, Fraile-Bermúdez AB, Alonso-Puyo J, Molano I, Rodriguez-Larrad A, Irazusta J. Serum Sestrin-1 Concentration Is Higher in Frail than Non-Frail Older People Living in Nursing Homes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031079. [PMID: 35162104 PMCID: PMC8834059 DOI: 10.3390/ijerph19031079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023]
Abstract
Given the increasing prevalence of frailty and its implications for public health, the identification of biomarkers to detect frailty is essential. Sestrin-1 is a protein with a protective role in muscle function. This study aimed to determine whether the serum sestrin-1 concentration differed between frail and non-frail populations and to investigate its association with frailty-related variables in 225 older women and men living in nursing homes (Gipuzkoa, Spain). Serum sestrin-1 concentration was measured by ELISA. Frailty, dependence, anthropometry, physical function, and physical activity were determined by validated tests and tools. The associations between sestrin-1 concentration and the other variables were determined using generalized linear models. The differences between frail and non-frail individuals were analyzed by the Mann–Whitney U-test, and receiver operating characteristic (ROC) curves were constructed to calculate the capability of sestrin-1 to detect frailty. Unexpectedly, frail individuals—according to the Fried Frailty Phenotype or the Clinical Frailty Scale—had higher serum sestrin-1 concentrations than non-frail individuals. Furthermore, the higher serum sestrin-1 concentration was associated with the increased frailty scores and dependence as well as the poorer physical function and the less physical activity. Given the contradictory results regarding serum sestrin-1 and frailty, further investigation is required to propose it as a molecular biomarker of frailty.
Collapse
Affiliation(s)
- Begoña Sanz
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
- Correspondence: ; Tel.: +34-946013307
| | - Chloe Rezola-Pardo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
| | - Haritz Arrieta
- Department of Nursing II, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 20014 Donostia-San Sebastián, Gipuzkoa, Spain;
| | - Ana Belén Fraile-Bermúdez
- Department of Nursing I, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain;
| | - Janire Alonso-Puyo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
| | - Irene Molano
- Residencia Sanmarcosene, Carretera de San Marcos, s/n, 20100 Errenteria, Gipuzkoa, Spain;
| | - Ana Rodriguez-Larrad
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Jon Irazusta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| |
Collapse
|
23
|
El Kharraz S, Dubois V, van Royen ME, Houtsmuller AB, Pavlova E, Atanassova N, Nguyen T, Voet A, Eerlings R, Handle F, Prekovic S, Smeets E, Moris L, Devlies W, Ohlsson C, Poutanen M, Verstrepen KJ, Carmeliet G, Launonen KM, Helminen L, Palvimo JJ, Libert C, Vanderschueren D, Helsen C, Claessens F. The androgen receptor depends on ligand-binding domain dimerization for transcriptional activation. EMBO Rep 2021; 22:e52764. [PMID: 34661369 DOI: 10.15252/embr.202152764] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 01/28/2023] Open
Abstract
Whereas dimerization of the DNA-binding domain of the androgen receptor (AR) plays an evident role in recognizing bipartite response elements, the contribution of the dimerization of the ligand-binding domain (LBD) to the correct functioning of the AR remains unclear. Here, we describe a mouse model with disrupted dimerization of the AR LBD (ARLmon/Y ). The disruptive effect of the mutation is demonstrated by the feminized phenotype, absence of male accessory sex glands, and strongly affected spermatogenesis, despite high circulating levels of testosterone. Testosterone replacement studies in orchidectomized mice demonstrate that androgen-regulated transcriptomes in ARLmon/Y mice are completely lost. The mutated AR still translocates to the nucleus and binds chromatin, but does not bind to specific AR binding sites. In vitro studies reveal that the mutation in the LBD dimer interface also affects other AR functions such as DNA binding, ligand binding, and co-regulator binding. In conclusion, LBD dimerization is crucial for the development of AR-dependent tissues through its role in transcriptional regulation in vivo. Our findings identify AR LBD dimerization as a possible target for AR inhibition.
Collapse
Affiliation(s)
- Sarah El Kharraz
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Vanessa Dubois
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | | | | | - Ekatarina Pavlova
- Institute of Experimental Morphology Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Nina Atanassova
- Institute of Experimental Morphology Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tien Nguyen
- Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Arnout Voet
- Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Roy Eerlings
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Florian Handle
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Prekovic
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elien Smeets
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lisa Moris
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Wout Devlies
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Gothenburg, Sweden
| | - Matti Poutanen
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Gothenburg, Sweden.,Research Centre for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Kevin J Verstrepen
- VIB Laboratory for Systems Biology and KU Leuven Laboratory for Genetics and Genomics, VIB - KU Leuven Center for Microbiology, Leuven, Belgium
| | - Geert Carmeliet
- Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | | | - Laura Helminen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Claude Libert
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Christine Helsen
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Ryan AS, Li G. Skeletal muscle myostatin gene expression and sarcopenia in overweight and obese middle‐aged and older adults. JCSM CLINICAL REPORTS 2021; 6:137-142. [PMID: 35311023 PMCID: PMC8932637 DOI: 10.1002/crt2.43] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Myostatin (MSTN) is a key negative regulator of muscle mass in humans and animals, having direct and indirect influences on molecular regulators of atrophy and hypertrophy, thus potentially impacting fitness and physical function. We have shown that myostatin is elevated in conditions of chronic disability (e.g. paretic limb of stroke). Our hypothesis is that myostatin would be elevated in older adults with sarcopenia. The purpose of this study was to examine the role of skeletal muscle myostatin in sarcopenia. Methods Sixty-four overweight to obese aged 45–81 years underwent a maximal aerobic capacity (VO2max) test, dual-energy X-ray absorptiometry (DXA) scan to determine appendicular lean tissue (ALM), and vastus lateralis muscle biopsy to determine myostatin mRNA expression by quantitative real time PCR (Q-RT-PCR). Rates of sarcopenia were determined using (ALM/BMI), and sarcopenia was defined as <0.789 in men and <0.512 in women. Subjects had low fitness (VO2max: 22.7 ± 0.7 mL/kg/min) and on average 40.9 ± 1% body fat. Results The prevalence of sarcopenia in this cohort was 16%. BMI, % body fat, and fat mass were higher in adults with sarcopenia than those without sarcopenia (all P < 0.001). Myostatin mRNA expression was lower in those without sarcopenia than those with sarcopenia (P < 0.05) and higher in men than women (P < 0.001). Myostatin expression was associated with BMI (r = 0.36, P < 0.01) and mid-thigh intramuscular fat (r = 0.29, P < 0.05). Conclusion Given that myostatin is important in muscle atrophy, fat accumulation, and sarcopenia, further work could address its implication in other aging cohorts of disability and chronic disease.
Collapse
Affiliation(s)
- Alice S. Ryan
- Department of Veterans Affairs, Department of Medicine, Division of Gerontology and Palliative Medicine, and the Baltimore VA Medical Center Geriatrics, Research, Education Center (GRECC) VA Maryland Health Care System Baltimore MD 21201 USA
| | - Guoyan Li
- Department of Veterans Affairs, Department of Medicine, Division of Gerontology and Palliative Medicine, and the Baltimore VA Medical Center Geriatrics, Research, Education Center (GRECC) VA Maryland Health Care System Baltimore MD 21201 USA
| |
Collapse
|
25
|
Fu S, Lin X, Yin L, Wang X. Androgen receptor regulates the proliferation of myoblasts under appropriate or excessive stretch through IGF-1 receptor mediated p38 and ERK1/2 pathways. Nutr Metab (Lond) 2021; 18:85. [PMID: 34526063 PMCID: PMC8444398 DOI: 10.1186/s12986-021-00610-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/21/2021] [Indexed: 12/23/2022] Open
Abstract
Background Androgen receptor (AR) exerts important roles in exercise-induced alterations of muscle mass, in which the proliferation and differentiation of satellite cells or myoblasts are crucial. Our previous study in C2C12 myoblasts demonstrated that 15% (mimic appropriate exercise) and 20% (mimic excessive exercise) stretches promoted and inhibited the proliferation respectively; and AR played a crucial role in 15% stretch-induced pro-proliferation through IGF-1-modulated PI3K/Akt, p38 and ERK1/2 pathways, but AR’s role in stretches-modulated proliferation of general myoblasts, especially 20% stretch, remains unclear, and the mechanisms need to be further clarified. Methods Firstly, the discrepancy in proliferation and the above indicators between L6 (without AR) and C2C12 (with AR) myoblasts were compared under 15% or 20% stretch. Then the influences of transfection AR or exogenous IGF-1 treatment on proliferation and these indicators were detected in stretched L6 myoblasts. Results (1) Under un-stretched state, the proliferation of L6 was slower than C2C12 cells. Furthermore, AR knockdown in C2C12 myoblasts repressed, while AR overexpression in L6 myoblasts promoted the proliferation. (2) 15% stretch-induced increases in the proliferation and activities of p38 and ERK1/2 were lower in L6 than C2C12 cells; AR overexpression enhanced the proliferation of 15% stretched L6 cells accompanied with the increases of p38 and ERK1/2 activities. (3) 20% stretch-induced anti-proliferation and inhibition of p38 activity were severer in L6 than C2C12 myoblasts; AR overexpression reversed the anti-proliferation of 20% stretch and enhanced p38 activity in L6 myoblasts. (4) In stretched L6 myoblasts, AR overexpression increased IGF-1R level despite no detectable IGF-1; and recombinant IGF-1 increased the proliferation, the level of IGF-1R, and the activities of p38 and ERK1/2 in 15% stretched L6 myoblasts. Conclusions The study demonstrated AR's crucial roles in stretches-regulated proliferation of myoblasts, and increased AR fulfilled 15% stretch's pro-proliferation via activating IGF-1R- p38 and ERK1/2 pathways while decreased AR achieved 20% stretch's anti-proliferation via inhibiting IGF-1R- p38 pathway, which is useful to understand in depth the role and mechanisms of AR in appropriate exercise increasing while excessive exercise decreasing muscle mass.
Collapse
Affiliation(s)
- Shaoting Fu
- Department of Exercise Physiology, School of Kinesiology, Shanghai University of Sport, 188 Hengren Road, Yangpu District, Shanghai, 200438, China.,Department of Kinesiology, College of Physical Education, Shanghai Normal University, Shanghai, 200234, China
| | - Xiaojing Lin
- Department of Exercise Physiology, School of Kinesiology, Shanghai University of Sport, 188 Hengren Road, Yangpu District, Shanghai, 200438, China
| | - Lijun Yin
- Department of Exercise Physiology, School of Kinesiology, Shanghai University of Sport, 188 Hengren Road, Yangpu District, Shanghai, 200438, China
| | - Xiaohui Wang
- Department of Exercise Physiology, School of Kinesiology, Shanghai University of Sport, 188 Hengren Road, Yangpu District, Shanghai, 200438, China.
| |
Collapse
|
26
|
Bhasin S, Hatfield DL, Hoffman JR, Kraemer WJ, Labotz M, Phillips SM, Ratamess NA. Anabolic-Androgenic Steroid Use in Sports, Health, and Society. Med Sci Sports Exerc 2021; 53:1778-1794. [PMID: 34261998 DOI: 10.1249/mss.0000000000002670] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This consensus statement is an update of the 1987 American College of Sports Medicine (ACSM) position stand on the use of anabolic-androgenic steroids (AAS). Substantial data have been collected since the previous position stand, and AAS use patterns have changed significantly. The ACSM acknowledges that lawful and ethical therapeutic use of AAS is now an accepted mainstream treatment for several clinical disorders; however, there is increased recognition that AAS are commonly used illicitly to enhance performance and appearance in several segments of the population, including competitive athletes. The illicit use of AAS by competitive athletes is contrary to the rules and ethics of many sport governing bodies. Thus, the ACSM deplores the illicit use of AAS for athletic and recreational purposes. This consensus statement provides a brief history of AAS use, an update on the science of how we now understand AAS to be working metabolically/biochemically, potential side effects, the prevalence of use among athletes, and the use of AAS in clinical scenarios.
Collapse
Affiliation(s)
- Shalender Bhasin
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Disa L Hatfield
- Department of Kinesiology, University of Rhode Island, Kingston, RI
| | - Jay R Hoffman
- Department of Physical Therapy, Ariel University, Ariel, Israel
| | - William J Kraemer
- Department of Human Sciences, The Ohio State University, Columbus, OH
| | | | | | - Nicholas A Ratamess
- Department of Health and Exercise Science, The College of New Jersey, Ewing, NJ
| |
Collapse
|
27
|
Sakakibara I, Yanagihara Y, Himori K, Yamada T, Sakai H, Sawada Y, Takahashi H, Saeki N, Hirakawa H, Yokoyama A, Fukada SI, Sawasaki T, Imai Y. Myofiber androgen receptor increases muscle strength mediated by a skeletal muscle splicing variant of Mylk4. iScience 2021; 24:102303. [PMID: 33870126 PMCID: PMC8041868 DOI: 10.1016/j.isci.2021.102303] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/16/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Androgens have a robust effect on skeletal muscles to increase muscle mass and strength. The molecular mechanism of androgen/androgen receptor (AR) action on muscle strength is still not well known, especially for the regulation of sarcomeric genes. In this study, we generated androgen-induced hypertrophic model mice, myofiber-specific androgen receptor knockout (cARKO) mice supplemented with dihydrotestosterone (DHT). DHT treatment increased grip strength in control mice but not in cARKO mice. Transcriptome analysis by RNA-seq, using skeletal muscles obtained from control and cARKO mice treated with or without DHT, identified a fast-type muscle-specific novel splicing variant of Myosin light-chain kinase 4 (Mylk4) as a target of AR in skeletal muscles. Mylk4 knockout mice exhibited decreased maximum isometric torque of plantar flexion and passive stiffness of myofibers due to reduced phosphorylation of Myomesin 1 protein. This study suggests that androgen-induced skeletal muscle strength is mediated with Mylk4 and Myomesin 1 axis. DHT increases muscle strength through myofiber AR Myofiber AR increases a fast-type muscle-specific novel splicing variant of Mylk4 MYLK4 regulates muscle strength and muscle stiffness MYLK4 induces phosphorylation of MYOM1
Collapse
Affiliation(s)
- Iori Sakakibara
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan.,Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan.,Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Yuta Yanagihara
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan.,Division of Laboratory Animal Research, Advanced Research Support Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Koichi Himori
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Hokkaido 060-8558, Japan
| | - Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Hokkaido 060-8558, Japan
| | - Hiroshi Sakai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan.,Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Yuichiro Sawada
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan.,Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Hirotaka Takahashi
- Division of Cell-Free Sciences, Proteo-Science Center (PROS), Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Noritaka Saeki
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan.,Division of Laboratory Animal Research, Advanced Research Support Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Hiroyuki Hirakawa
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University (TMDU), Graduate School, Tokyo 113-8510, Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tatsuya Sawasaki
- Division of Cell-Free Sciences, Proteo-Science Center (PROS), Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan.,Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan.,Division of Laboratory Animal Research, Advanced Research Support Center, Ehime University, Toon, Ehime 791-0295, Japan.,Research Unit for Skeletal Health and Diseases, Ehime University, Toon, Ehime 791-0295, Japan
| |
Collapse
|
28
|
Gharahdaghi N, Phillips BE, Szewczyk NJ, Smith K, Wilkinson DJ, Atherton PJ. Links Between Testosterone, Oestrogen, and the Growth Hormone/Insulin-Like Growth Factor Axis and Resistance Exercise Muscle Adaptations. Front Physiol 2021; 11:621226. [PMID: 33519525 PMCID: PMC7844366 DOI: 10.3389/fphys.2020.621226] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
Maintenance of skeletal muscle mass throughout the life course is key for the regulation of health, with physical activity a critical component of this, in part, due to its influence upon key hormones such as testosterone, estrogen, growth hormone (GH), and insulin-like growth factor (IGF). Despite the importance of these hormones for the regulation of skeletal muscle mass in response to different types of exercise, their interaction with the processes controlling muscle mass remain unclear. This review presents evidence on the importance of these hormones in the regulation of skeletal muscle mass and their responses, and involvement in muscle adaptation to resistance exercise. Highlighting the key role testosterone plays as a primary anabolic hormone in muscle adaptation following exercise training, through its interaction with anabolic signaling pathways and other hormones via the androgen receptor (AR), this review also describes the potential importance of fluctuations in other hormones such as GH and IGF-1 in concert with dietary amino acid availability; and the role of estrogen, under the influence of the menstrual cycle and menopause, being especially important in adaptive exercise responses in women. Finally, the downstream mechanisms by which these hormones impact regulation of muscle protein turnover (synthesis and breakdown), and thus muscle mass are discussed. Advances in our understanding of hormones that impact protein turnover throughout life offers great relevance, not just for athletes, but also for the general and clinical populations alike.
Collapse
Affiliation(s)
| | | | | | | | - Daniel J. Wilkinson
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research and Nottingham National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, United Kingdom
| | - Philip J. Atherton
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research and Nottingham National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, United Kingdom
| |
Collapse
|
29
|
Priego T, Martín AI, González-Hedström D, Granado M, López-Calderón A. Role of hormones in sarcopenia. VITAMINS AND HORMONES 2021; 115:535-570. [PMID: 33706961 DOI: 10.1016/bs.vh.2020.12.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aging involves numerous changes in body composition that include a decrease in skeletal muscle mass. The gradual reduction in muscle mass is associated with a simultaneous decrease in muscle strength, which leads to reduced mobility, fragility and loss of independence. This process called sarcopenia is secondary to several factors such as sedentary lifestyle, inadequate nutrition, chronic inflammatory state and neurological alterations. However, the endocrine changes associated with aging seem to be of special importance in the development of sarcopenia. On one hand, advancing age is associated with a decreased secretion of the main hormones that stimulate skeletal muscle mass and function (growth hormone, insulin-like growth factor 1 (IGFI), testosterone and estradiol). On the other hand, the alteration of the IGF-I signaling along with decreased insulin sensitivity also have an important impact on myogenesis. Other hormones that decline with aging such as the adrenal-derived dehydroepiandrosterone, thyroid hormones and vitamin D seem to also be involved in sarcopenia. Adipokines released by adipose tissue show important changes during aging and can affect muscle physiology and metabolism. In addition, catabolic hormones such as cortisol and angiotensin II can accelerate aged-induced muscle atrophy, as they are involved in muscle wasting and their levels increase with age. The role played by all of these hormones and the possible use of some of them as therapeutic tools for treating sarcopenia will be discussed.
Collapse
Affiliation(s)
- T Priego
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - A I Martín
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - D González-Hedström
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; Pharmactive Biotech Products S.L. Parque Científico de Madrid. Avenida del Doctor Severo Ochoa, 37 Local 4J, 28108 Alcobendas, Madrid, Spain
| | - M Granado
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición. Instituto de Salud Carlos III, Madrid, Spain
| | - A López-Calderón
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
30
|
Sur S, Chaturvedi K, Sharma A, Malik S, Rani S, Kumar V. Ambient temperature affects multiple drivers of physiology and behaviour: adaptation for timely departure of obligate spring migrants. J Exp Biol 2020; 223:jeb236109. [PMID: 33161378 DOI: 10.1242/jeb.236109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/02/2020] [Indexed: 12/26/2022]
Abstract
We investigated the role of ambient temperature in departure from wintering areas of migratory black-headed buntings in spring. Birds transferred at 22 and 35°C to long days were compared with one another and with controls held on short days for indices of readiness to migrate (Zugunruhe, fattening, mass gain), levels of testosterone and gonadal recrudescence. Temperature affected the development of migratory behaviour and physiology: buntings under long days at 35°C, compared with those at 22°C, showed altered migratory behaviour (daily activity and Zugunruhe onset), and enhanced muscle growth and plasma testosterone levels, but showed no effect on testis growth. Temperature was perceived at both peripheral and central levels, and affected multiple molecular drivers culminating into the migratory phenotype. This was evidenced by post-mortem comparison of the expression of 13 genes with known functions in the skin (temperature-sensitive TRP channels: trpv4 and trpm8), hypothalamus and/or midbrain (migration-linked genes: th, ddc, adcyap1 and vps13a) and flight muscles (muscle growth associated genes: ar, srd5a3, pvalb, mtor, myod, mstn and hif1a). In photostimulated birds, the expression of trpv4 in skin, th in the hypothalamus and midbrain, and srd5a3, ar, pvalb and mtor in flight muscle, in parallel with testosterone levels, was greater at 35°C than at 22°C. These results demonstrate the role of ambient temperature in development of the spring migration phenotype, and suggest that transcriptional responsiveness to temperature is a component of the overall adaptive strategy in latitudinal songbird migrants for timely departure from wintering areas in spring.
Collapse
Affiliation(s)
- Sayantan Sur
- Department of Zoology, University of Delhi, Delhi 110007, India
| | | | - Aakansha Sharma
- Department of Zoology, University of Delhi, Delhi 110007, India
| | - Shalie Malik
- Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Sangeeta Rani
- Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Vinod Kumar
- Department of Zoology, University of Delhi, Delhi 110007, India
| |
Collapse
|
31
|
Aquila G, Re Cecconi AD, Forti M, Frapolli R, Bello E, Novelli D, Russo I, Licandro SA, Staszewsky L, Martinelli GB, Talamini L, Pasetto L, Resovi A, Giavazzi R, Scanziani E, Careccia G, Vénéreau E, Masson S, Latini R, D’Incalci M, Piccirillo R. Trabectedin and Lurbinectedin Extend Survival of Mice Bearing C26 Colon Adenocarcinoma, without Affecting Tumor Growth or Cachexia. Cancers (Basel) 2020; 12:cancers12082312. [PMID: 32824440 PMCID: PMC7463843 DOI: 10.3390/cancers12082312] [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: 07/13/2020] [Revised: 07/31/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
Trabectedin (ET743) and lurbinectedin (PM01183) limit the production of inflammatory cytokines that are elevated during cancer cachexia. Mice carrying C26 colon adenocarcinoma display cachexia (i.e., premature death and body wasting with muscle, fat and cardiac tissue depletion), high levels of inflammatory cytokines and subsequent splenomegaly. We tested whether such drugs protected these mice from cachexia. Ten-week-old mice were inoculated with C26 cells and three days later randomized to receive intravenously vehicle or 0.05 mg/kg ET743 or 0.07 mg/kg PM01183, three times a week for three weeks. ET743 or PM01183 extended the lifespan of C26-mice by 30% or 85%, respectively, without affecting tumor growth or food intake. Within 13 days from C26 implant, both drugs did not protect fat, muscle and heart from cachexia. Since PM01183 extended the animal survival more than ET743, we analyzed PM01183 further. In tibialis anterior of C26-mice, but not in atrophying myotubes, PM01183 restrained the NF-κB/PAX7/myogenin axis, possibly reducing the pro-inflammatory milieu, and failed to limit the C/EBPβ/atrogin-1 axis. Inflammation-mediated splenomegaly of C26-mice was inhibited by PM01183 for as long as the treatment lasted, without reducing IL-6, M-CSF or IL-1β in plasma. ET743 and PM01183 extend the survival of C26-bearing mice unchanging tumor growth or cachexia but possibly restrain muscle-related inflammation and C26-induced splenomegaly.
Collapse
Affiliation(s)
- Giorgio Aquila
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.); (M.F.); (G.B.M.)
| | - Andrea David Re Cecconi
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.); (M.F.); (G.B.M.)
| | - Mara Forti
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.); (M.F.); (G.B.M.)
| | - Roberta Frapolli
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (R.F.); (E.B.); (S.A.L.); (R.G.); (M.D.)
| | - Ezia Bello
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (R.F.); (E.B.); (S.A.L.); (R.G.); (M.D.)
| | - Deborah Novelli
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (D.N.); (I.R.); (L.S.); (S.M.); (R.L.)
| | - Ilaria Russo
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (D.N.); (I.R.); (L.S.); (S.M.); (R.L.)
| | - Simonetta Andrea Licandro
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (R.F.); (E.B.); (S.A.L.); (R.G.); (M.D.)
| | - Lidia Staszewsky
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (D.N.); (I.R.); (L.S.); (S.M.); (R.L.)
| | - Giulia Benedetta Martinelli
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.); (M.F.); (G.B.M.)
| | - Laura Talamini
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (L.T.); (L.P.)
| | - Laura Pasetto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (L.T.); (L.P.)
| | - Andrea Resovi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy;
| | - Raffaella Giavazzi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (R.F.); (E.B.); (S.A.L.); (R.G.); (M.D.)
| | - Eugenio Scanziani
- Dipartimento di Medicina Veterinaria, Università di Milano, 20133 Milan, Italy;
- Mouse and Animal Pathology Lab (MAPLab), Fondazione UniMi, Università di Milano, 20139 Milan, Italy
| | - Giorgia Careccia
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.C.); (E.V.)
| | - Emilie Vénéreau
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (G.C.); (E.V.)
| | - Serge Masson
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (D.N.); (I.R.); (L.S.); (S.M.); (R.L.)
| | - Roberto Latini
- Department of Cardiovascular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (D.N.); (I.R.); (L.S.); (S.M.); (R.L.)
| | - Maurizio D’Incalci
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (R.F.); (E.B.); (S.A.L.); (R.G.); (M.D.)
| | - Rosanna Piccirillo
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, 20156 Milan, Italy; (G.A.); (A.D.R.C.); (M.F.); (G.B.M.)
- Correspondence: ; Tel.: +39-02-39014371
| |
Collapse
|
32
|
Androgen receptor in satellite cells is not essential for muscle regenerations. EXPERIMENTAL RESULTS 2020. [DOI: 10.1017/exp.2020.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AbstractThe anabolic effects of androgen on skeletal muscles are thought to be mediated by androgen receptor (AR). Although multiple studies concerning the effects of AR in males have been performed, the molecular mechanisms of AR in skeletal muscles remain unclear. Here we first confirmed that satellite cells from mouse hindlimb muscles express AR. We then generated satellite cell-specific AR knockout mice using Pax7CreERT2 and ARL2/Y mice to test whether AR in satellite cells is necessary for muscle regeneration. Surprisingly, we found that muscle regeneration was compromised in both Pax7CreERT2(Fan)/+ control mice and Pax7CreERT2(Fan)/+;ARL2/Y mice compared to ARL2/Y mice. However, Pax7CreERT2(Gaka)/+;ARL2/Y;R26tdTomato/+ mice showed no significant differences between control and mutant muscle regeneration. These findings indicate that AR in satellite cells is not essential for muscle regeneration. We propose that Pax7CreERT2(Fan)/+ control mice should be included in all experiments, because these mice negatively affect the muscle regeneration and show the mild regeneration phenotype.
Collapse
|
33
|
Primary osteoporosis in men: an unmet medical need. Fertil Steril 2020; 112:791-798. [PMID: 31731933 DOI: 10.1016/j.fertnstert.2019.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/27/2022]
Abstract
Osteoporosis is a skeletal disease characterized by loss of bone strength and increased risk of fractures. Even though fracture prevalence is higher in women, fractures also constitute a significant public health issue in older men. Men are screened less and more frequently undertreated than female patients. It is the goal of this review, to summarize updated information about the current understanding of pathophysiology and clinical aspects of diagnosis and treatment of osteoporosis in men.
Collapse
|
34
|
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
|
35
|
Cui M, Gang X, Wang G, Xiao X, Li Z, Jiang Z, Wang G. A cross-sectional study: Associations between sarcopenia and clinical characteristics of patients with type 2 diabetes. Medicine (Baltimore) 2020; 99:e18708. [PMID: 31914078 PMCID: PMC6959879 DOI: 10.1097/md.0000000000018708] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sarcopenia is a geriatric syndrome and it impairs physical function. Patients with type 2 diabetes mellitus (T2DM) are at a higher risk of sarcopenia. The purpose of this study is to explore characteristics of general information and metabolic factors of sarcopenia in patients with T2DM in the northeast of China, and provide information for the prevention and treatment of sarcopenia in clinical practice.Patients with T2DM aged ≥65 were recruited in Changchun from March 2017 to February 2018. Questionnaires of general information, physical examination, laboratory and imaging examination were conducted. The patients were assigned into sarcopenia group and non-sarcopenia group according to the diagnostic criteria proposed by Asian working group for sarcopenia (AWGS), and the differences between 2 groups were analyzed.A total of 132 participants were included in this study, of which, 38 (28.8%) were diagnosed with sarcopenia. 94 (71.2%) were with no sarcopenia. Logistic regression analysis showed that age (OR: 1.182, 95%CI: 1.038-1.346), trunk fat mass (TFM) (OR: 1.499, 95%CI: 1.146-1.960) and free thyroxine (FT4) (OR: 1.342, 95%CI: 1.102-1.635) were independent risk factors for sarcopenia. BMI (body mass index) (OR: 0.365, 95%CI: 0.236-0.661), exercise (OR: 0.016, 95%CI: 0.001-0.169), female (OR: 0.000, 95%CI: 0.00-0.012), metformin (OR: 0.159, 95%CI: 0.026-0.967) and TSM (trunk skeletal muscle mass) (OR: 0.395, 95%CI: 0.236-0.661) were protective factors for sarcopenia.Sarcopenia in patients with T2DM is associated with increased age, increased TFM and increased FT4 level. Regular exercise, female, metformin administrations, high BMI and increased TSM are associated with lower risk of sarcopenia.
Collapse
|
36
|
Kraemer WJ, Ratamess NA, Hymer WC, Nindl BC, Fragala MS. Growth Hormone(s), Testosterone, Insulin-Like Growth Factors, and Cortisol: Roles and Integration for Cellular Development and Growth With Exercise. Front Endocrinol (Lausanne) 2020; 11:33. [PMID: 32158429 PMCID: PMC7052063 DOI: 10.3389/fendo.2020.00033] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
Hormones are largely responsible for the integrated communication of several physiological systems responsible for modulating cellular growth and development. Although the specific hormonal influence must be considered within the context of the entire endocrine system and its relationship with other physiological systems, three key hormones are considered the "anabolic giants" in cellular growth and repair: testosterone, the growth hormone superfamily, and the insulin-like growth factor (IGF) superfamily. In addition to these anabolic hormones, glucocorticoids, mainly cortisol must also be considered because of their profound opposing influence on human skeletal muscle anabolism in many instances. This review presents emerging research on: (1) Testosterone signaling pathways, responses, and adaptations to resistance training; (2) Growth hormone: presents new complexity with exercise stress; (3) Current perspectives on IGF-I and physiological adaptations and complexity these hormones as related to training; and (4) Glucocorticoid roles in integrated communication for anabolic/catabolic signaling. Specifically, the review describes (1) Testosterone as the primary anabolic hormone, with an anabolic influence largely dictated primarily by genomic and possible non-genomic signaling, satellite cell activation, interaction with other anabolic signaling pathways, upregulation or downregulation of the androgen receptor, and potential roles in co-activators and transcriptional activity; (2) Differential influences of growth hormones depending on the "type" of the hormone being assayed and the magnitude of the physiological stress; (3) The exquisite regulation of IGF-1 by a family of binding proteins (IGFBPs 1-6), which can either stimulate or inhibit biological action depending on binding; and (4) Circadian patterning and newly discovered variants of glucocorticoid isoforms largely dictating glucocorticoid sensitivity and catabolic, muscle sparing, or pathological influence. The downstream integrated anabolic and catabolic mechanisms of these hormones not only affect the ability of skeletal muscle to generate force; they also have implications for pharmaceutical treatments, aging, and prevalent chronic conditions such as metabolic syndrome, insulin resistance, and hypertension. Thus, advances in our understanding of hormones that impact anabolic: catabolic processes have relevance for athletes and the general population, alike.
Collapse
Affiliation(s)
- William J. Kraemer
- Department of Human Sciences, The Ohio State University, Columbus, OH, United States
- *Correspondence: William J. Kraemer
| | - Nicholas A. Ratamess
- Department of Health and Exercise Science, The College of New Jersey, Ewing, NJ, United States
| | - Wesley C. Hymer
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States
| | - Bradley C. Nindl
- Department of Sports Medicine, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | | |
Collapse
|
37
|
Rosa-Caldwell ME, Greene NP. Muscle metabolism and atrophy: let's talk about sex. Biol Sex Differ 2019; 10:43. [PMID: 31462271 PMCID: PMC6714453 DOI: 10.1186/s13293-019-0257-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 08/16/2019] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle health is a strong predictor of overall health and longevity. Pathologies affecting skeletal muscle such as cancer cachexia, intensive care unit treatment, muscular dystrophies, and others are associated with decreased quality of life and increased mortality. Recent research has begun to determine that these muscular pathologies appear to present and develop differently between males and females. However, to our knowledge, there has yet to be a comprehensive review on musculoskeletal differences between males and females and how these differences may contribute to sex differences in muscle pathologies. Herein, we present a review of the current literature on muscle phenotype and physiology between males and females and how these differences may contribute to differential responses to atrophic stimuli. In general, females appear to be more susceptible to disuse induced muscle wasting, yet protected from inflammation induced (such as cancer cachexia) muscle wasting compared to males. These differences may be due in part to differences in muscle protein turnover, satellite cell content and proliferation, hormonal interactions, and mitochondrial differences between males and females. However, more works specifically examining muscle pathologies in females are necessary to more fully understand the inherent sex-based differences in muscle pathologies between the sexes and how they may correspond to different clinical treatments.
Collapse
Affiliation(s)
- Megan E Rosa-Caldwell
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Nicholas P Greene
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
38
|
Son BK, Eto M, Oura M, Ishida Y, Taniguchi S, Ito K, Umeda-Kameyama Y, Kojima T, Akishita M. Low-Intensity Exercise Suppresses CCAAT/Enhancer-Binding Protein δ/Myostatin Pathway Through Androgen Receptor in Muscle Cells. Gerontology 2019; 65:397-406. [PMID: 31096217 DOI: 10.1159/000499826] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 03/24/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Androgen production following exercise has been suggested to contribute anabolic actions of muscle. However, the underlying mechanisms of the androgen receptor (AR) in androgen's action are still unclear. OBJECTIVE In the present study, we examined androgen/AR-mediated action in exercise, especially for the suppression of myostatin, a potent negative regulator of muscle mass. METHODS To examine the effects of exercise, we employed low-intensity exercise in mice and electric pulse stimulation (EPS) in C2C12 myotubes. Androgen production by C2C12 myotubes was measured by enzyme-linked immunosorbent assay. To block the action of AR, we pretreated C2C12 myotubes with flutamide. Quantitative real-time polymerase chain reaction was used to determine the expression levels of proteolytic genes including CCAAT/enhancer-binding protein delta (C/EBPδ), myostatin and muscle E3 ubiquitin ligases, as well as myogenic genes such as myogenin and PGC1α. The activation of 5'-adenosine-activated protein kinase and STAT3 was determined by Western blot analysis. RESULTS Both mRNA and protein levels of AR significantly increased in skeletal muscle of low-intensity exercised mice and C2C12 myotubes exposed to EPS. Production of testosterone and dihydrotestosterone from EPS-treated C2C12 myotubes was markedly increased. Of interest, we found that myostatin was clearly inhibited by EPS, and its inhibition was significantly abrogated when AR was blocked by flutamide. To test how AR suppresses myostatin, we examined the effects of EPS on C/EBPδ because the promoter region of myostatin has several C/EBP recognition sites. C/EBPδ expression was decreased by EPS, and this decrease was negated by flutamide. IL-6 and phospho-STAT3 (pSTAT3) expression, the downstream pathway of myostatin, were decreased by EPS and this was also reversed by flutamide. Similar downregulation of C/EBPδ, myostatin, and IL-6 was seen in skeletal muscle of low-intensity exercised mice. CONCLUSIONS Muscle AR expression and androgen production were increased by exercise and EPS treatment. As a mechanistical insight, it is suggested that AR inhibited myostatin expression transcriptionally by C/EBPδ suppression, which negatively influences IL-6/pSTAT3 expression and consequently contributes to the prevention of muscle proteolysis during exercise.
Collapse
Affiliation(s)
- Bo-Kyung Son
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan, .,Institute of Gerontology, The University of Tokyo, Tokyo, Japan,
| | - Masato Eto
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Miya Oura
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Ishida
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakiko Taniguchi
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Koichi Ito
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yumi Umeda-Kameyama
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taro Kojima
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
39
|
Clarke MV, Russell PK, Zajac JD, Davey RA. The androgen receptor in the hypothalamus positively regulates hind-limb muscle mass and voluntary physical activity in adult male mice. J Steroid Biochem Mol Biol 2019; 189:187-194. [PMID: 30853652 DOI: 10.1016/j.jsbmb.2019.02.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 10/27/2022]
Abstract
We have previously shown that expression of the androgen receptor (AR) in neurons within the brain positively regulates hind-limb muscle mass and physical activity in male mice. To further investigate the region of the brain responsible for mediating these effects of testosterone and to determine whether they are only important for muscle mass accrual during development or whether they are also important for the maintenance of muscle mass in the adult, we deleted the AR specifically in the hypothalamus of adult male mice (Hyp-ARKOs). Hyp-ARKO mice were generated by bilateral stereotaxic microinjection of an adeno-associated virus (AAV) expressing GFP and iCre recombinase under the control of the e-synapsin promoter into the hypothalamus of 10-week-old exon 3-AR floxed male mice. AR mRNA was deleted by 45% in the hypothalamus of Hyp-ARKOs at 5 weeks post-AAV-eSyn-iCre injection. This led to an increase in the mass of the androgen-dependent organs, seminal vesicles and kidneys, by 30% (P < 0.01) and 10% (P < 0.05) respectively, and an increase in serum luteinizing hormone (LH) by 2 fold (P < 0.05). Whilst the mean value for serum testosterone was higher in the Hyp-ARKOs, this did not reach statistical significance. Despite a phenotype consistent with increased androgen bioactivity in Hyp-ARKOs, which would be expected to increase muscle mass, the mass of the hind-limb muscles, gastrocnemius (Gast) (P = 0.001), extensor digitorum longus (EDL) (P < 0.001) and soleus (Sol) (P < 0.01) were paradoxically decreased by 12-19% compared to controls. Voluntary physical activity was reduced by 65% (P < 0.05) in Hyp-ARKO male mice and was associated with a reduction in gene expression of Drd1a and Maob (P ≤ 0.05) in the hypothalamus, suggesting involvement of the brain dopaminergic system. These data provide compelling evidence that androgen signalling via the AR in the hypothalamus acts to positively regulate the maintenance of hind-limb muscle mass and voluntary activity in adult male mice, independent of AR signalling in peripheral tissues.
Collapse
Affiliation(s)
- Michele V Clarke
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia
| | - Patricia K Russell
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia
| | - Rachel A Davey
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia.
| |
Collapse
|
40
|
Martignani E, Miretti S, Vincenti L, Baratta M. Correlation between estrogen plasma level and miRNAs in muscle of Piedmontese cattle. Domest Anim Endocrinol 2019; 67:37-41. [PMID: 30690256 DOI: 10.1016/j.domaniend.2018.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/21/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022]
Abstract
A loss-of-function mutation of the myostatin gene has a very high prevalence in the Piedmontese cattle breed. The effect of such mutation is a double-muscle phenotype because of hypertrophy. However, differences in muscle mass development can still be detected in individuals of this breed. Such differences must be generated by other factors controlling skeletal muscle development. MicroRNAs are short noncoding RNA molecules that modulate gene expression at a post-transcriptional level. MicroRNAs have been demonstrated to be involved in skeletal muscle development, and some of them are controlled by steroid hormone signaling. Data on estrogen signaling are lacking, whereas more studies have been carried out on the effect of androgens. We aimed at identifying putative estrogen responsive miRNAs that might be involved in skeletal muscle development. At a slaughterhouse, we collected muscle samples from longissimus dorsi and blood samples. Blood 17β-estradiol concentration was assessed, and RNA was extracted from muscle samples. The animals we sampled were divided into groups according to estrogen blood concentration, and through qPCR expression, levels of 7 muscle-related miRNAs were evaluated. We found that miR-26b (P < 0.01), miR-27a-5p (P < 0.05), miR-27b (P < 0.05), and miR-199a-3p (P < 0.01) were differentially expressed among experimental groups. Expression levels of miR-26b were reduced approximately 50% in samples with a low blood estrogen concentrations, and the other miRNAs showed a tendency to increase their expression levels when blood estrogen levels were higher. The variations of the circulating concentrations of estrogen in Piedmontese cattle might influence muscle mass development through miRNAs and thus contribute to individual variability in a breed with a high prevalence of a myostatin point mutation.
Collapse
Affiliation(s)
- E Martignani
- Department of Veterinary Science, University of Torino, Largo Braccini 2, 10095 Grugliasco, TO, Italy.
| | - S Miretti
- Department of Veterinary Science, University of Torino, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| | - L Vincenti
- Department of Veterinary Science, University of Torino, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| | - M Baratta
- Department of Veterinary Science, University of Torino, Largo Braccini 2, 10095 Grugliasco, TO, Italy
| |
Collapse
|
41
|
Laurent M, Dupont J, Dejaeger M, Gielen E. Myostatin: A Powerful Biomarker for Sarcopenia and Frailty? Gerontology 2019; 65:383-384. [DOI: 10.1159/000495839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/28/2018] [Indexed: 01/11/2023] Open
|
42
|
Jardí F, Kim N, Laurent MR, Khalil R, Deboel L, Schollaert D, van Lenthe GH, Decallonne B, Carmeliet G, Claessens F, Vanderschueren D. Androgen Receptor in Neurons Slows Age-Related Cortical Thinning in Male Mice. J Bone Miner Res 2019; 34:508-519. [PMID: 30496619 DOI: 10.1002/jbmr.3625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022]
Abstract
Androgens via the androgen receptor (AR) are required for optimal male bone health. The target cell(s) for the effects of androgens on cortical bone remain(s) incompletely understood. In females, estrogen receptor alpha in neurons is a negative regulator of cortical and trabecular bone. Whether neuronal AR regulates bone mass in males remains unexplored. Here, we inactivated AR in neurons using a tamoxifen-inducible CreERT2 under the control of the neuronal promoter Thy1. Tamoxifen induced a 70% to 80% reduction of AR mRNA levels in Thy1-CreERT2-positive brain regions cerebral cortex and brainstem as well as in the peripheral nervous tissue of male neuronal AR knockout (N-ARKO) mice. Hypothalamic AR mRNA levels were only marginally reduced and the hypothalamic-pituitary-gonadal axis remained unaffected, as determined by normal levels of serum testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH). In contrast to orchidectomy, deletion of neuronal AR did not alter body weight, body composition, hindlimb muscle mass, grip strength, or wheel running. MicroCT analysis of the femur revealed no changes in bone accrual during growth in N-ARKO mice. However, 36- and 46-week-old N-ARKO mice displayed an accelerated age-related cortical involution, namely a more pronounced loss of cortical thickness and strength, which occurred in the setting of androgen sufficiency. Neuronal AR inactivation decreased the cancellous bone volume fraction in L5 vertebra but not in the appendicular skeleton of aging mice. MicroCT findings were corroborated in the tibia and after normalization of hormonal levels. Serum markers of bone turnover and histomorphometry parameters were comparable between genotypes, except for a 30% increase in osteoclast surface in the trabecular compartment of 36-week-old N-ARKO mice. Cortical bone loss in N-ARKO mice was associated with an upregulation of Ucp1 expression in brown adipose tissue, a widely used readout for sympathetic tone. We conclude that androgens preserve cortical integrity in aging male mice via AR in neurons. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Nari Kim
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Michaël R Laurent
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,Gerontology and Geriatrics, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Rougin Khalil
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Ludo Deboel
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Dieter Schollaert
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - G H van Lenthe
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Aging (CHROMETA), KU Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Seo JY, Kim JH, Kong YY. Unraveling the Paradoxical Action of Androgens on Muscle Stem Cells. Mol Cells 2019; 42:97-103. [PMID: 30759971 PMCID: PMC6399011 DOI: 10.14348/molcells.2019.0004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 12/25/2022] Open
Abstract
Androgens act in almost all tissues throughout the lifetime and have important roles in skeletal muscles. The levels of androgens increase during puberty and remain sustained at high levels in adulthood. Because androgens have an anabolic effect on skeletal muscles and muscle stem cells, these increased levels of androgens after puberty should lead to spontaneous muscle hypertrophy and hyperplasia in adulthood. However, the maintenance of muscle volume, myonuclei number per myofiber, and quiescent state of satellite cells in adulthood despite the high levels of androgens produces paradoxical outcomes. Our recent study revealed that the physiological increase of androgens at puberty initiates the transition of muscle stem cells from proliferation to quiescence by the androgen-Mindbomb1-Notch signaling axis. This newly discovered androgen action on skeletal muscles underscores the physiological importance of androgens on muscle homeostasis throughout life. This review will provide an overview of the new androgen action on skeletal muscles and discuss the paradoxical effects of androgens suggested in previous studies.
Collapse
Affiliation(s)
- Ji-Yun Seo
- School of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| | - Ji-Hoon Kim
- School of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| | - Young-Yun Kong
- School of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| |
Collapse
|
44
|
Paul RG, McMahon CD, Elston MS, Conaglen JV. GH replacement titrated to serum IGF-1 does not reduce concentrations of myostatin in blood or skeletal muscle. Growth Horm IGF Res 2019; 44:11-16. [PMID: 30543929 DOI: 10.1016/j.ghir.2018.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Traditional weight-based regimens of GH replacement are more effective at reversing the loss of skeletal muscle in GH-deficient adults than currently recommended regimens, where the dose of GH is increased to restore serum concentrations of IGF-1. While weight-based regimens increase concentrations of IGF-1 and decrease concentrations of myostatin, it is not known whether the reduced effectiveness of individually titrated GH regimens is due to ongoing hypersecretion of myostatin. Consequently, the aims of this study were to determine whether concentrations of myostatin in blood and skeletal muscle are increased in GH-deficient adults, and whether these concentrations are decreased by GH replacement regimens titrated to restore serum IGF-1. DESIGN Twenty-six GH deficient adults (18 men and 8 women) were treated with individualised regimens of recombinant human GH aiming to achieve serum concentrations of IGF-1 within one standard deviation of the age- and gender-adjusted mean. Plasma concentrations of myostatin were measured at baseline and after 6 months of treatment were compared to fifteen healthy controls (9 men and 6 women). Skeletal muscle biopsies were performed in 19 of these GH-deficient adults (15 men and 4 women) and 10 of the healthy controls (6 men and 4 women). Expression of IGF-1 and myostatin mRNA was determined by qPCR. RESULTS Concentrations of IGF-1 in serum and mRNA in skeletal muscle were reduced, and concentrations of myostatin in plasma and mRNA in skeletal muscle were increased in GH-deficient adults at baseline (P < .05 versus healthy controls). Despite restoring concentrations of IGF-1, GH replacement did not reduce concentrations of myostatin in either blood or skeletal muscle. Concentrations of IGF-1 and myostatin in both blood and skeletal muscle were positively correlated in GH-deficient adults at baseline (P < .05), but not in GH-replete adults. CONCLUSIONS Concentrations of myostatin in blood and skeletal muscle are increased in GH-deficient adults. Despite normalising concentrations of IGF-1, individualised regimens of GH replacement do not reduce concentrations of myostatin in blood or skeletal muscle. Ongoing hypersecretion of myostatin may explain why individually titrated GH replacement regimens are less effective than higher weight-based regimens in increasing skeletal muscle mass.
Collapse
Affiliation(s)
- Ryan G Paul
- AgResearch Ltd, Ruakura Research Centre, Private Bag 3123, Hamilton, New Zealand; Waikato Clinical Campus, University of Auckland, Private Bag 3200, Hamilton 3240, New Zealand.
| | - Chris D McMahon
- AgResearch Ltd, Ruakura Research Centre, Private Bag 3123, Hamilton, New Zealand
| | - Marianne S Elston
- Waikato Clinical Campus, University of Auckland, Private Bag 3200, Hamilton 3240, New Zealand
| | - John V Conaglen
- Waikato Clinical Campus, University of Auckland, Private Bag 3200, Hamilton 3240, New Zealand
| |
Collapse
|
45
|
Age-related bone loss and sarcopenia in men. Maturitas 2019; 122:51-56. [PMID: 30797530 DOI: 10.1016/j.maturitas.2019.01.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 12/20/2022]
Abstract
Bone and muscle are required for mobility but they also have endocrine and metabolic functions. In ageing as well as in many chronic diseases, bone loss and muscle atrophy occur simultaneously, leading to concomitant osteoporosis and sarcopenia. This occurs in both genders but compared with postmenopausal women, men appear to be better protected against age-related bone and muscle decay. Sex steroids (both androgens like testosterone and oestrogens like estradiol) are mainly responsible for musculoskeletal sexual dimorphism. They stimulate peak bone and muscle mass accretion during puberty and midlife, and prevent subsequent loss in ageing men but not post-menopausal women. Still, recent studies have highlighted the importance of intrinsic ageing mechanisms such as cellular senescence and oxidative stress in both genders. Sarcopenia may predispose to dysmobility, frailty, falls and fractures, but whether so-called osteosarcopenia qualifies as a distinct entity remains debated. Although randomized clinical trials in male osteoporosis are smaller and therefore underpowered for some outcomes like hip fractures, the available evidence suggests that the clinical diagnostic and therapeutic approach to male osteoporosis is largely similar to that in postmenopausal women. There is a clear unmet medical need for effective and safe anabolic drugs to rebuild the ageing skeleton, muscle, and preferably both tissues simultaneously. The Wnt/sclerostin and myostatin/activin receptor signalling pathways appear particularly promising in this regard. In this narrative review, we aim to provide an overview of our current understanding of the pathophysiology and treatment of male osteoporosis and sarcopenia, and interactions between these two diseases.
Collapse
|
46
|
Arrieta H, Hervás G, Rezola-Pardo C, Ruiz-Litago F, Iturburu M, Yanguas JJ, Gil SM, Rodriguez-Larrad A, Irazusta J. Serum Myostatin Levels Are Higher in Fitter, More Active, and Non-Frail Long-Term Nursing Home Residents and Increase after a Physical Exercise Intervention. Gerontology 2018; 65:229-239. [PMID: 30463070 DOI: 10.1159/000494137] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/01/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Myostatin has been proposed as a candidate biomarker for frailty and sarcopenia. However, the relationship of myostatin with these conditions remains inconclusive. OBJECTIVE To determine the association of serum myostatin concentration with body composition, physical fitness, physical activity level, and frailty in long-term nursing home residents. We also aimed to ascertain the effect of an exercise program on myostatin levels. METHODS We obtained study data on 112 participants from long-term nursing homes. Participants were randomly assigned to a control or an intervention group and performed a 6-month multicomponent exercise program. Serum myostatin levels were analyzed by ELISA. Assessments also included body composition (anthropometry and bioelectrical impedance), physical fitness (Senior Fitness Test), physical activity level (accelerometry), and frailty (Fried frailty criteria, Clinical Frailty Scale, and Tilburg frailty indicator). RESULTS The concentration of myostatin at baseline was positively correlated with: a leaner body composition (p < 0.05), and a higher number of steps per day and light and moderate-vigorous physical activity in women (p < 0.005); greater upper and lower limb strength, endurance, and poorer flexibility (p < 0.05) in men; and better performance (less time) in the 8-ft timed up-and-go test in both women (p < 0.01) and men (p < 0.005). We observed higher concentrations of serum myostatin in non-frail than in frail participants (p < 0.05). Additionally, we found that the implemented physical exercise intervention, which was effective to improve physical fitness, increased myostatin concentration in men (p < 0.05) but not in women. The improvements in physical condition were related with increases in serum myostatin only in men (p < 0.05-0.01). CONCLUSIONS Higher serum levels of myostatin were found to be associated with better physical fitness. The improvements in physical fitness after the intervention were positively related to increases in myostatin concentrations in men. These results seem to rule out the idea that high serum myostatin levels are indicative of frailty in long-term nursing home residents. However, although the direction of association was opposite to that expected for the function of myostatin, the use of this protein as a biomarker for physical fitness, rather than frailty, merits further study.
Collapse
Affiliation(s)
- Haritz Arrieta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain,
| | - Gotzone Hervás
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Chloe Rezola-Pardo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Fátima Ruiz-Litago
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | | | - Susana María Gil
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Ana Rodriguez-Larrad
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jon Irazusta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
47
|
Gibson DA, Simitsidellis I, Collins F, Saunders PTK. Endometrial Intracrinology: Oestrogens, Androgens and Endometrial Disorders. Int J Mol Sci 2018; 19:E3276. [PMID: 30360364 PMCID: PMC6214123 DOI: 10.3390/ijms19103276] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/05/2018] [Accepted: 10/15/2018] [Indexed: 12/26/2022] Open
Abstract
Peripheral tissue metabolism of steroids (intracrinology) is now accepted as a key way in which tissues, such as the endometrium, can utilise inactive steroids present in the blood to respond to local physiological demands and 'fine-tune' the activation or inhibition of steroid hormone receptor-dependent processes. Expression of enzymes that play a critical role in the activation and inactivation of bioactive oestrogens (E1, E2) and androgens (A4, T, DHT), as well as expression of steroid hormone receptors, has been detected in endometrial tissues and cells recovered during the menstrual cycle. There is robust evidence that increased expression of aromatase is important for creating a local microenvironment that can support a pregnancy. Measurement of intra-tissue concentrations of steroids using liquid chromatography⁻tandem mass spectrometry has been important in advancing our understanding of a role for androgens in the endometrium, acting both as active ligands for the androgen receptor and as substrates for oestrogen biosynthesis. The emergence of intracrinology, associated with disordered expression of key enzymes such as aromatase, in the aetiology of common women's health disorders such as endometriosis and endometrial cancer has prompted renewed interest in the development of drugs targeting these pathways, opening up new opportunities for targeted therapies and precision medicine.
Collapse
Affiliation(s)
- Douglas A Gibson
- Centre for Inflammation Research, The University of Edinburgh, EH16 4TJ Edinburgh, UK.
| | - Ioannis Simitsidellis
- Centre for Inflammation Research, The University of Edinburgh, EH16 4TJ Edinburgh, UK.
| | - Frances Collins
- Centre for Inflammation Research, The University of Edinburgh, EH16 4TJ Edinburgh, UK.
| | - Philippa T K Saunders
- Centre for Inflammation Research, The University of Edinburgh, EH16 4TJ Edinburgh, UK.
| |
Collapse
|
48
|
Handelsman DJ, Hirschberg AL, Bermon S. Circulating Testosterone as the Hormonal Basis of Sex Differences in Athletic Performance. Endocr Rev 2018; 39:803-829. [PMID: 30010735 PMCID: PMC6391653 DOI: 10.1210/er.2018-00020] [Citation(s) in RCA: 272] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/18/2018] [Indexed: 12/22/2022]
Abstract
Elite athletic competitions have separate male and female events due to men's physical advantages in strength, speed, and endurance so that a protected female category with objective entry criteria is required. Prior to puberty, there is no sex difference in circulating testosterone concentrations or athletic performance, but from puberty onward a clear sex difference in athletic performance emerges as circulating testosterone concentrations rise in men because testes produce 30 times more testosterone than before puberty with circulating testosterone exceeding 15-fold that of women at any age. There is a wide sex difference in circulating testosterone concentrations and a reproducible dose-response relationship between circulating testosterone and muscle mass and strength as well as circulating hemoglobin in both men and women. These dichotomies largely account for the sex differences in muscle mass and strength and circulating hemoglobin levels that result in at least an 8% to 12% ergogenic advantage in men. Suppression of elevated circulating testosterone of hyperandrogenic athletes results in negative effects on performance, which are reversed when suppression ceases. Based on the nonoverlapping, bimodal distribution of circulating testosterone concentration (measured by liquid chromatography-mass spectrometry)-and making an allowance for women with mild hyperandrogenism, notably women with polycystic ovary syndrome (who are overrepresented in elite athletics)-the appropriate eligibility criterion for female athletic events should be a circulating testosterone of <5.0 nmol/L. This would include all women other than those with untreated hyperandrogenic disorders of sexual development and noncompliant male-to-female transgender as well as testosterone-treated female-to-male transgender or androgen dopers.
Collapse
Affiliation(s)
- David J Handelsman
- ANZAC Research Institute, University of Sydney, Concord, New South Wales, Australia.,Department of Andrology, Concord Hospital, Sydney, New South Wales, Australia
| | - Angelica L Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Stephane Bermon
- Laboratoire Motricité Humaine, Education, Sport, Santé, Université Côte d'Azur, Nice, France.,Health and Science Department, International Association of Athletics Federations, Monaco
| |
Collapse
|
49
|
Jardí F, Laurent MR, Dubois V, Kim N, Khalil R, Decallonne B, Vanderschueren D, Claessens F. Androgen and estrogen actions on male physical activity: a story beyond muscle. J Endocrinol 2018; 238:R31-R52. [PMID: 29743340 DOI: 10.1530/joe-18-0125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/09/2018] [Indexed: 12/15/2022]
Abstract
Physical inactivity is a pandemic that contributes to several chronic diseases and poses a significant burden on health care systems worldwide. The search for effective strategies to combat sedentary behavior has led to an intensification of the research efforts to unravel the biological substrate controlling activity. A wide body of preclinical evidence makes a strong case for sex steroids regulating physical activity in both genders, albeit the mechanisms implicated remain unclear. The beneficial effects of androgens on muscle as well as on other peripheral functions might play a role in favoring adaptation to exercise. Alternatively or in addition, sex steroids could act on specific brain circuitries to boost physical activity. This review critically discusses the evidence supporting a role for androgens and estrogens stimulating male physical activity, with special emphasis on the possible role of peripheral and/or central mechanisms. Finally, the potential translation of these findings to humans is briefly discussed.
Collapse
Affiliation(s)
- Ferran Jardí
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Michaël R Laurent
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Gerontology and GeriatricsDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Vanessa Dubois
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nari Kim
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Rougin Khalil
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Brigitte Decallonne
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental EndocrinologyDepartment of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Molecular Endocrinology LaboratoryDepartment of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Harada N. Role of androgens in energy metabolism affecting on body composition, metabolic syndrome, type 2 diabetes, cardiovascular disease, and longevity: lessons from a meta-analysis and rodent studies. Biosci Biotechnol Biochem 2018; 82:1667-1682. [PMID: 29957125 DOI: 10.1080/09168451.2018.1490172] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Testosterone is a sex hormone produced by testicular Leydig cells in males. Blood testosterone concentrations increase at three time-periods in male life-fetal, neonatal (which can be separated into newborn and infant periods), and pubertal stages. After peaking in the early 20s, the blood bioactive testosterone level declines by 1-2% each year. It is increasingly apparent that a low testosterone level impairs general physical and mental health in men. Here, this review summarizes recent systematic reviews and meta-analyses of epidemiological studies in males (including cross-sectional, longitudinal, and androgen deprivation studies, and randomized controlled testosterone replacement trials) in relation to testosterone and obesity, body composition, metabolic syndrome, type 2 diabetes, cardiovascular disease, and longevity. Furthermore, underlying mechanisms are discussed using data from rodent studies involving castration or androgen receptor knockout. This review provides an update understanding of the role of testosterone in energy metabolism. Abbreviations AR: androgen receptor; CV: cardiovascular; FDA: US Food and Drug Administration; HFD: high-fat diet; KO: knockout; MetS: metabolic syndrome; RCT: randomized controlled trial; SHBG: sex hormone binding globulin; SRMA: systematic review and meta-analysis; TRT: testosterone replacement therapy; T2DM:type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Naoki Harada
- a Division of Applied Life Sciences , Graduate School of Life and Environmental Sciences, Osaka Prefecture University , Sakai , Osaka , Japan
| |
Collapse
|