17α-estradiol Alleviates High-Fat Diet-Induced Inflammatory and Metabolic Dysfunction in Skeletal Muscle of Male and Female Mice

Skeletal muscle has a central role in maintaining metabolic homeostasis. 17α-estradiol (17α-E2), a naturally-occurring non-feminizing diastereomer of 17β-estradiol that demonstrates efficacy for improving metabolic outcomes in male, but not female, mice. Despite several lines of evidence showing that 17α-E2 treatment improves metabolic parameters in middle-aged obese and old male mice through effects in brain, liver, and white adipose tissue little is known about how 17α-E2 alters skeletal muscle metabolism, and what role this may play in mitigating metabolic declines. Therefore, this study aimed to determine if 17α-E2 treatment improves metabolic outcomes in skeletal muscle from obese male and female mice following chronic high fat diet (HFD) administration. We hypothesized that male, but not female, mice, would benefit from 17α-E2 treatment during HFD. To test this hypothesis, we used a multi-omics approach to determine changes in lipotoxic lipid intermediates, metabolites, and proteins related to metabolic homeostasis. In male mice, we show that 17α-E2 alleviates HFD-induced metabolic detriments of skeletal muscle by reducing the accumulation of diacylglycerol (DAGs) and ceramides, inflammatory cytokine levels, and reduced the abundance of most of the proteins related to lipolysis and beta-oxidation. In contrast to males, 17α-E2 treatment in female mice had little effect on the DAGs and ceramides content, muscle inflammatory cytokine levels, or changes to the relative abundance of proteins involved in beta-oxidation. These data support to the growing evidence that 17α-E2 treatment could be beneficial for overall metabolic health in male mammals.

[1]  Roshini Sathiaseelan,et al.  Metabolic benefits of 17α-estradiol in liver are partially mediated by ERβ in male mice , 2023, bioRxiv.

[2]  S. Austad,et al.  A Genetically Heterogeneous Rat Model with Divergent Mitochondrial Genomes , 2023, The journals of gerontology. Series A, Biological sciences and medical sciences.

[3]  T. Collares,et al.  Mild calorie restriction, but not 17α-estradiol, extends ovarian reserve and fertility in female mice , 2022, Experimental Gerontology.

[4]  Patrick D. Shipman,et al.  Determining the contributions of protein synthesis and breakdown to muscle atrophy requires non‐steady‐state equations , 2021, Journal of cachexia, sarcopenia and muscle.

[5]  Richard A. Miller,et al.  17-a-estradiol has sex-specific effects on neuroinflammation that are partly reversed by gonadectomy. , 2021, The journals of gerontology. Series A, Biological sciences and medical sciences.

[6]  Ricardo Aurino Pinho,et al.  Distinct mechanisms involving diacylglycerol, ceramides, and inflammation underlie insulin resistance in oxidative and glycolytic muscles from high fat-fed rats , 2021, Scientific Reports.

[7]  N. Rosenthal,et al.  17‐a‐estradiol late in life extends lifespan in aging UM‐HET3 male mice; nicotinamide riboside and three other drugs do not affect lifespan in either sex , 2021, Aging cell.

[8]  Roshini Sathiaseelan,et al.  17α-Estradiol prevents ovariectomy-mediated obesity and bone loss , 2020, Experimental Gerontology.

[9]  Roshini Sathiaseelan,et al.  17α-estradiol modulates IGF1 and hepatic gene expression in a sex-specific manner. , 2020, The journals of gerontology. Series A, Biological sciences and medical sciences.

[10]  J. C. Price,et al.  Sex differences in changes of protein synthesis with rapamycin treatment are minimized when metformin is added to rapamycin , 2020, GeroScience.

[11]  W. Freeman,et al.  Health benefits attributed to 17α-estradiol, a lifespan-extending compound, are mediated through estrogen receptor α , 2020, bioRxiv.

[12]  J. C. Price,et al.  CORES OF REPRODUCIBILITY IN PHYSIOLOGY: THE USE OF DEUTERATED WATER FOR THE MEASUREMENT OF PROTEIN SYNTHESIS. , 2020, Journal of applied physiology.

[13]  Benjamin F. Miller,et al.  CORP: The use of deuterated water for the measurement of protein synthesis , 2020 .

[14]  B. Miller,et al.  Massage as a mechanotherapy promotes skeletal muscle protein and ribosomal turnover but does not mitigate muscle atrophy during disuse in adult rats , 2020, Acta physiologica.

[15]  B. Miller,et al.  Mitochondrial adaptations to exercise do not require Bcl2‐mediated autophagy but occur with BNIP3/Parkin activation , 2020, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[16]  A. Hevener,et al.  The Impact of Skeletal Muscle ERα on Mitochondrial Function and Metabolic Health. , 2020, Endocrinology.

[17]  A. Bartke,et al.  17α-Estradiol promotes ovarian aging in growth hormone receptor knockout mice, but not wild-type littermates , 2019, Experimental Gerontology.

[18]  B. Miller,et al.  Muscle‐specific changes in protein synthesis with aging and reloading after disuse atrophy , 2019, Journal of cachexia, sarcopenia and muscle.

[19]  J. C. Price,et al.  Short-term calorie restriction and 17α-estradiol administration elicit divergent effects on proteostatic processes and protein content in metabolically active tissues. , 2019, The journals of gerontology. Series A, Biological sciences and medical sciences.

[20]  Tzulip Phang,et al.  Intermuscular adipose tissue directly modulates skeletal muscle insulin sensitivity in humans. , 2019, American journal of physiology. Endocrinology and metabolism.

[21]  O. Fiehn,et al.  17‐α estradiol ameliorates age‐associated sarcopenia and improves late‐life physical function in male mice but not in females or castrated males , 2019, Aging cell.

[22]  T. Griffin,et al.  GC–MS metabolic profiling reveals fructose-2,6-bisphosphate regulates branched chain amino acid metabolism in the heart during fasting , 2019, Metabolomics.

[23]  T. Griffin,et al.  GC–MS metabolic profiling reveals fructose-2,6-bisphosphate regulates branched chain amino acid metabolism in the heart during fasting , 2019, Metabolomics.

[24]  S. Dasari,et al.  Remodeling of skeletal muscle mitochondrial proteome with high-fat diet involves greater changes to β-oxidation than electron transfer proteins in mice. , 2018, American journal of physiology. Endocrinology and metabolism.

[25]  A. Galecki,et al.  Male lifespan extension with 17‐α estradiol is linked to a sex‐specific metabolomic response modulated by gonadal hormones in mice , 2018, Aging cell.

[26]  E. Ravussin,et al.  Skeletal muscle ceramides and daily fat oxidation in obesity and diabetes. , 2018, Metabolism: clinical and experimental.

[27]  M. Garratt,et al.  Hormone actions controlling sex-specific life-extension , 2018, Aging.

[28]  M. Low,et al.  17α‐estradiol acts through hypothalamic pro‐opiomelanocortin expressing neurons to reduce feeding behavior , 2017, Aging cell.

[29]  L. Harrison-Bernard,et al.  High fat diet consumption differentially affects adipose tissue inflammation and adipocyte size in obesity-prone and obesity-resistant rats , 2017, International Journal of Obesity.

[30]  B. Miller,et al.  Long-term rates of mitochondrial protein synthesis are increased in mouse skeletal muscle with high-fat feeding regardless of insulin-sensitizing treatment. , 2017, American journal of physiology. Endocrinology and metabolism.

[31]  Richard A. Miller,et al.  Sex differences in lifespan extension with acarbose and 17‐α estradiol: gonadal hormones underlie male‐specific improvements in glucose tolerance and mTORC2 signaling , 2017, Aging cell.

[32]  B. Miller,et al.  Influence of Nrf2 activators on subcellular skeletal muscle protein and DNA synthesis rates after 6 weeks of milk protein feeding in older adults , 2017, GeroScience.

[33]  Dudley Lamming,et al.  Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an α‐glucosidase inhibitor or a Nrf2‐inducer , 2016, Aging cell.

[34]  A. Hevener,et al.  Skeletal muscle action of estrogen receptor α is critical for the maintenance of mitochondrial function and metabolic homeostasis in females , 2016, Science Translational Medicine.

[35]  Shuo Lin,et al.  Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism , 2016, Skeletal Muscle.

[36]  G. Shulman,et al.  17α-Estradiol Alleviates Age-related Metabolic and Inflammatory Dysfunction in Male Mice Without Inducing Feminization , 2016, The journals of gerontology. Series A, Biological sciences and medical sciences.

[37]  B. Ames,et al.  Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males , 2013, Aging cell.

[38]  Christophe Ley,et al.  Detecting outliers: Do not use standard deviation around the mean, use absolute deviation around the median , 2013 .

[39]  U. Eriksson,et al.  Imaging of neutral lipids by oil red O for analyzing the metabolic status in health and disease , 2013, Nature Protocols.

[40]  M. Bruss,et al.  Calorie restriction does not increase short-term or long-term protein synthesis. , 2013, The journals of gerontology. Series A, Biological sciences and medical sciences.

[41]  W. Niu,et al.  The role of AMPK/mTOR/S6K1 signaling axis in mediating the physiological process of exercise-induced insulin sensitization in skeletal muscle of C57BL/6 mice. , 2012, Biochimica et biophysica acta.

[42]  A. Jamurtas,et al.  The effects of aging, physical training, and a single bout of exercise on mitochondrial protein expression in human skeletal muscle , 2012, Experimental Gerontology.

[43]  B. Viollet,et al.  Defining the Contribution of AMP-activated Protein Kinase (AMPK) and Protein Kinase C (PKC) in Regulation of Glucose Uptake by Metformin in Skeletal Muscle Cells* , 2012, The Journal of Biological Chemistry.

[44]  M. Bruss,et al.  A comprehensive assessment of mitochondrial protein synthesis and cellular proliferation with age and caloric restriction , 2012, Aging cell.

[45]  B. Bergman,et al.  Localisation and composition of skeletal muscle diacylglycerol predicts insulin resistance in humans , 2012, Diabetologia.

[46]  Peter J. Chomentowski,et al.  Skeletal Muscle Triglycerides, Diacylglycerols, and Ceramides in Insulin Resistance , 2011, Diabetes.

[47]  M. Belury,et al.  Hepatic steatosis by dietary-conjugated linoleic acid is accompanied by accumulation of diacylglycerol and increased membrane-associated protein kinase C ε in mice. , 2011, Molecular nutrition & food research.

[48]  Z. Varghese,et al.  Inflammatory stress exacerbates ectopic lipid deposition in C57BL/6J mice , 2011, Lipids in Health and Disease.

[49]  M. Jensen,et al.  Sex‐ and Depot‐Dependent Differences in Adipogenesis in Normal‐Weight Humans , 2010, Obesity.

[50]  Cherith Moore,et al.  Motivation and diabetes self-management , 2010, Chronic illness.

[51]  E. Masoro Caloric restriction-induced life extension of rats and mice: a critique of proposed mechanisms. , 2009, Biochimica et biophysica acta.

[52]  V. Procaccio,et al.  Mitochondrial Effects of Estrogen Are Mediated by Estrogen Receptor α in Brain Endothelial Cells , 2008, Journal of Pharmacology and Experimental Therapeutics.

[53]  J. Bienkowska,et al.  Estrogen Receptors α and β Mediate Distinct Pathways of Vascular Gene Expression, Including Genes Involved in Mitochondrial Electron Transport and Generation of Reactive Oxygen Species , 2007 .

[54]  S. Walrand,et al.  Enhanced Muscle Mixed and Mitochondrial Protein Synthesis Rates after a High‐fat or High‐sucrose Diet , 2007, Obesity.

[55]  David W. Russell,et al.  LMSD: LIPID MAPS structure database , 2006, Nucleic Acids Res..

[56]  B. Pedersen,et al.  Influence of TNF-α and IL-6 infusions on insulin sensitivity and expression of IL-18 in humans , 2006 .

[57]  C. Leeuwenburgh,et al.  Caloric restriction in humans: Potential pitfalls and health concerns , 2006, Mechanisms of Ageing and Development.

[58]  B. Goodpaster,et al.  Skeletal muscle lipid accumulation in obesity, insulin resistance, and type 2 diabetes , 2004, Pediatric diabetes.

[59]  A. Namane,et al.  Protein sequencing and identification using tandem mass spectrometry. Edited by Michael Kinter, Nicholas E. Sherman, published by Wiley-Interscience Series on Mass Spectrometry, 2000, 301 p. , 2002 .

[60]  Margaret S. Wu,et al.  Role of AMP-activated protein kinase in mechanism of metformin action. , 2001, The Journal of clinical investigation.

[61]  Rena R Wing,et al.  Skeletal muscle fatty acid metabolism in association with insulin resistance, obesity, and weight loss. , 1999, American journal of physiology. Endocrinology and metabolism.

[62]  P. Zimmet,et al.  Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus. Provisional report of a WHO Consultation , 1998, Diabetic medicine : a journal of the British Diabetic Association.

[63]  G Dailey,et al.  Metabolic effects of metformin in non-insulin-dependent diabetes mellitus. , 1995, The New England journal of medicine.

[64]  H. Yki-Järvinen,et al.  Kinetics of glucose disposal in whole body and across the forearm in man. , 1987, The Journal of clinical investigation.

[65]  C. Bailey,et al.  Effect of metformin on glucose metabolism in mouse soleus muscle. , 1986, Diabete & metabolisme.

[66]  R. DeFronzo,et al.  The Effect of Insulin on the Disposal of Intravenous Glucose: Results from Indirect Calorimetry and Hepatic and Femoral Venous Catheterization , 1981, Diabetes.

[67]  J. Folch,et al.  A simple method for the isolation and purification of total lipides from animal tissues. , 1957, The Journal of biological chemistry.

[68]  B. Bergman,et al.  HPLC-MS/MS Methods for Diacylglycerol and Sphingolipid Molecular Species in Skeletal Muscle. , 2019, Methods in molecular biology.

[69]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[70]  Olga Ilkayeva,et al.  Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. , 2008, Cell metabolism.

[71]  Thomas D. Schmittgen,et al.  Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2 2 DD C T Method , 2022 .