Skip to main content
Top
Published in: Endocrine 3/2015

01-04-2015 | Original Article

Sex-related differences in the effects of high-fat diets on DHEA-treated rats

Authors: Ana Lúcia Cecconello, Marcia Trapp, Ana Lúcia Hoefel, Cláudia Vieira Marques, Bruno Dutra Arbo, Gabriela Osterkamp, Luiz Carlos Rios Kucharski, Maria Flávia Marques Ribeiro

Published in: Endocrine | Issue 3/2015

Login to get access

Abstract

Several studies have investigated the beneficial effects of dehydroepiandrosterone (DHEA) on lipid and glucose metabolism. However, many of these studies are inconclusive about the effects of DHEA administration on metabolic disorders, and there appear to be sex-related differences in the effects of DHEA treatment. Few animal studies have addressed the effects of DHEA on diet-induced metabolic disorders. The present study sought to ascertain whether sex differences exist in the effects of a high-fat diet (HFD) on weight gain, adiposity, and biochemical and hormonal parameters in DHEA-treated rats. Rats were fed a HFD for 4 weeks and simultaneously received treatment with DHEA (10 mg/kg by subcutaneous injection) once weekly. Body weight, retroperitoneal fat depot weight, serum glucose, insulin, and leptin levels, and hepatic lipids were measured. HFD exposure increased the adiposity index in both sexes, the hepatic triglyceride content in both sexes, and the hepatic total cholesterol level in males. Moreover, the HFD induced an increase in blood glucose levels in both sexes, and hyperinsulinemia in males. In this experimental model, DHEA treatment reduced hepatic triglyceride levels only in females, regardless of HFD exposure. Exposure to a HFD, even if it does not cause obesity, may enhance risk factors for metabolic disorders, and males are more sensitive to this effect. DHEA treatment can help prevent metabolic derangements, but its effect varies with sex.
Literature
1.
go back to reference N. Hariri, L. Thibault, High-fat diet-induced obesity in animal models. Nutr. Res. Rev. 23, 270–299 (2010)CrossRefPubMed N. Hariri, L. Thibault, High-fat diet-induced obesity in animal models. Nutr. Res. Rev. 23, 270–299 (2010)CrossRefPubMed
3.
go back to reference M. Bergman, Inadequacies of current approaches to prediabetes and diabetes prevention. Endocrine 44, 623–633 (2013)CrossRefPubMed M. Bergman, Inadequacies of current approaches to prediabetes and diabetes prevention. Endocrine 44, 623–633 (2013)CrossRefPubMed
4.
go back to reference T. Priego, J. Sánchez, C. Picó, A. Palou, Sex-differential expression of metabolism- related genes in response to a high-fat diet. Obesity 16, 819–826 (2008)CrossRefPubMed T. Priego, J. Sánchez, C. Picó, A. Palou, Sex-differential expression of metabolism- related genes in response to a high-fat diet. Obesity 16, 819–826 (2008)CrossRefPubMed
5.
go back to reference O.D. Taraschenko, I.M. Maisonneuve, S.D. Glick, Sex differences in high fat-induced obesity in rats: effects of 18-methoxycoronaridine. Physiol. Behav. 103, 308–314 (2011)CrossRefPubMed O.D. Taraschenko, I.M. Maisonneuve, S.D. Glick, Sex differences in high fat-induced obesity in rats: effects of 18-methoxycoronaridine. Physiol. Behav. 103, 308–314 (2011)CrossRefPubMed
6.
go back to reference K. Michalakis, G. Mintziori, A. Kaprara, B.C. Tarlatzis, D.G. Goulis, The complex interaction between obesity, metabolic syndrome and reproductive axis: a narrative review. Metabolism 62, 457–478 (2013)CrossRefPubMed K. Michalakis, G. Mintziori, A. Kaprara, B.C. Tarlatzis, D.G. Goulis, The complex interaction between obesity, metabolic syndrome and reproductive axis: a narrative review. Metabolism 62, 457–478 (2013)CrossRefPubMed
7.
go back to reference J. Avruch, Insulin signal transduction through protein kinase cascades. Mol. Cell. Biochem. 182, 31–48 (1998)CrossRefPubMed J. Avruch, Insulin signal transduction through protein kinase cascades. Mol. Cell. Biochem. 182, 31–48 (1998)CrossRefPubMed
8.
go back to reference A.R. Saltiel, J.E. Pessin, Insulin signaling pathways in time and space. Trends Cell Biol. 12, 65–71 (2002)CrossRefPubMed A.R. Saltiel, J.E. Pessin, Insulin signaling pathways in time and space. Trends Cell Biol. 12, 65–71 (2002)CrossRefPubMed
9.
go back to reference R.V. Farese, M.P. Sajan, M.L. Standaert, Insulin-sensitive protein kinases (atypical protein kinase C and protein kinase B/Akt): actions and defects in obesity and type II diabetes. Exp. Biol. Med. 230, 593–605 (2005) R.V. Farese, M.P. Sajan, M.L. Standaert, Insulin-sensitive protein kinases (atypical protein kinase C and protein kinase B/Akt): actions and defects in obesity and type II diabetes. Exp. Biol. Med. 230, 593–605 (2005)
10.
go back to reference C.A. Lalli, J.R. Pauli, P.O. Prada, D.E. Cintra, E.R. Ropelle, L.A. Velloso, M.J. Saad, Statin modulates insulin signaling and insulin resistance in liver and muscle of rats fed a high-fat diet. Metabolism 57, 57–65 (2008)CrossRefPubMed C.A. Lalli, J.R. Pauli, P.O. Prada, D.E. Cintra, E.R. Ropelle, L.A. Velloso, M.J. Saad, Statin modulates insulin signaling and insulin resistance in liver and muscle of rats fed a high-fat diet. Metabolism 57, 57–65 (2008)CrossRefPubMed
11.
go back to reference G.D. Ferreira, A. Germeyer, A. de Barros Machado, T.L. do Nascimento, T. Strowitzki, I.S. Brum, H. Von Eye Corleta, E. Capp, Metformin modulates PI3K and GLUT4 expression and Akt/PKB phosphorylation in human endometrial stromal cells after stimulation with androgen and insulin. Eur. J. Obstet. Gynecol. Reprod. Biol. 175, 157–162 (2014)CrossRefPubMed G.D. Ferreira, A. Germeyer, A. de Barros Machado, T.L. do Nascimento, T. Strowitzki, I.S. Brum, H. Von Eye Corleta, E. Capp, Metformin modulates PI3K and GLUT4 expression and Akt/PKB phosphorylation in human endometrial stromal cells after stimulation with androgen and insulin. Eur. J. Obstet. Gynecol. Reprod. Biol. 175, 157–162 (2014)CrossRefPubMed
12.
go back to reference J. Karbowska, Z. Kochan, Fat-reducing effects of dehydroepiandrosterone involve upregulation of ATGL and HSL expression, and stimulation of lipolysis in adipose tissue. Steroids 77, 1359–1365 (2012)CrossRefPubMed J. Karbowska, Z. Kochan, Fat-reducing effects of dehydroepiandrosterone involve upregulation of ATGL and HSL expression, and stimulation of lipolysis in adipose tissue. Steroids 77, 1359–1365 (2012)CrossRefPubMed
13.
go back to reference N. Orentreich, J.L. Brind, J.H. Vogelman, R. Andres, H. Baldwin, Long-term longitudinal measurements of plasma dehydroepiandrosterone sulfate in normal men. J. Clin. Endocrinol. Metab. 75, 1002–1004 (1992)PubMed N. Orentreich, J.L. Brind, J.H. Vogelman, R. Andres, H. Baldwin, Long-term longitudinal measurements of plasma dehydroepiandrosterone sulfate in normal men. J. Clin. Endocrinol. Metab. 75, 1002–1004 (1992)PubMed
14.
go back to reference J. Sánchez, F. Pérez-Heredia, T. Priego, M.P. Portillo, S. Zamora, M. Garaulet, A. Palou, Dehydroepiandrosterone prevents age-associated alterations, increasing insulin sensitivity. J. Nutr. Biochem. 19, 809–818 (2008)CrossRefPubMed J. Sánchez, F. Pérez-Heredia, T. Priego, M.P. Portillo, S. Zamora, M. Garaulet, A. Palou, Dehydroepiandrosterone prevents age-associated alterations, increasing insulin sensitivity. J. Nutr. Biochem. 19, 809–818 (2008)CrossRefPubMed
15.
go back to reference M. Garaulet, F. Pérex-Llamas, T. Fuente, S. Zamora, F.J. Tebar, Anthropometric, computed tomography and fat cell data in an obese population: relationship with insulin, leptin, tumor necrosis factor-alpha, sex hormone-binding globulin and sex hormones. Eur. J. Endocrinol. 143, 657–666 (2000)CrossRefPubMed M. Garaulet, F. Pérex-Llamas, T. Fuente, S. Zamora, F.J. Tebar, Anthropometric, computed tomography and fat cell data in an obese population: relationship with insulin, leptin, tumor necrosis factor-alpha, sex hormone-binding globulin and sex hormones. Eur. J. Endocrinol. 143, 657–666 (2000)CrossRefPubMed
16.
go back to reference M.K. Shelby, D.J. Crouch, D.L. Black, T.A. Robert, R. Heltsley, Screening indicators of dehydroepiandosterone, androstenedione, and dihydrotestosterone use: a literature review. J. Anal. Toxicol. 35, 638–655 (2011)CrossRefPubMed M.K. Shelby, D.J. Crouch, D.L. Black, T.A. Robert, R. Heltsley, Screening indicators of dehydroepiandosterone, androstenedione, and dihydrotestosterone use: a literature review. J. Anal. Toxicol. 35, 638–655 (2011)CrossRefPubMed
17.
go back to reference C. Gómez-Santos, J.J. Hernández-Morante, F.J. Tébar, E. Granero, M. Garaulet, Differential effect of oral dehydroepiandrosterone-sulphate on metabolic syndrome features in pre- and postmenopausal obese women. Clin. Endocrinol. 77, 548–554 (2012)CrossRef C. Gómez-Santos, J.J. Hernández-Morante, F.J. Tébar, E. Granero, M. Garaulet, Differential effect of oral dehydroepiandrosterone-sulphate on metabolic syndrome features in pre- and postmenopausal obese women. Clin. Endocrinol. 77, 548–554 (2012)CrossRef
18.
go back to reference P.A. Hansen, D.H. Han, L.A. Nolte, M. Chen, J.O. Holloszy, DHEA protects against visceral obesity and muscle insulin resistance in rats fed a high-fat diet. Am. J. Physiol. 273, R1704–R1708 (1997)PubMed P.A. Hansen, D.H. Han, L.A. Nolte, M. Chen, J.O. Holloszy, DHEA protects against visceral obesity and muscle insulin resistance in rats fed a high-fat diet. Am. J. Physiol. 273, R1704–R1708 (1997)PubMed
19.
go back to reference Z. Magyar, G. Bekesi, K. Racz, J. Feher, Z. Schaff, G. Lengyel, A. Blazovics, G. Illyes, D. Szombath, A. Hrabak, B. Szekacs, P. Gergics, I. Marczell, E. Dinya, J. Rigo Jr, Z. Tulassay, Increased total scavenger capacity and decreased liver fat content in rats fed dehydroepiandrosterone and its sulphate on a high-fat diet. Gerontology 57, 343–349 (2011)CrossRefPubMed Z. Magyar, G. Bekesi, K. Racz, J. Feher, Z. Schaff, G. Lengyel, A. Blazovics, G. Illyes, D. Szombath, A. Hrabak, B. Szekacs, P. Gergics, I. Marczell, E. Dinya, J. Rigo Jr, Z. Tulassay, Increased total scavenger capacity and decreased liver fat content in rats fed dehydroepiandrosterone and its sulphate on a high-fat diet. Gerontology 57, 343–349 (2011)CrossRefPubMed
20.
go back to reference C.S. Campbell, L.C. Caperuto, A.E. Hirata, E.P. Araujo, L.A. Velloso, M.J. Saad, C.R. Carvalho, The phosphatidylinositol/AKT/atypical PKC pathway is involved in the improved insulin sensitivity by DHEA in muscle and liver of rats in vivo. Life Sci. 76, 57–70 (2004)CrossRefPubMed C.S. Campbell, L.C. Caperuto, A.E. Hirata, E.P. Araujo, L.A. Velloso, M.J. Saad, C.R. Carvalho, The phosphatidylinositol/AKT/atypical PKC pathway is involved in the improved insulin sensitivity by DHEA in muscle and liver of rats in vivo. Life Sci. 76, 57–70 (2004)CrossRefPubMed
21.
go back to reference M. Aragno, R. Mastrocola, G. Alloatti, I. Vercellinatto, P. Bardini, S. Geuna, M.G. Catalano, O. Danni, G. Boccuzzi, Oxidative stress triggers cardiac fibrosis in the heart of diabetic rats. Endocrinology 149, 380–388 (2008)CrossRefPubMed M. Aragno, R. Mastrocola, G. Alloatti, I. Vercellinatto, P. Bardini, S. Geuna, M.G. Catalano, O. Danni, G. Boccuzzi, Oxidative stress triggers cardiac fibrosis in the heart of diabetic rats. Endocrinology 149, 380–388 (2008)CrossRefPubMed
22.
go back to reference J.J. Hernández-Morante, F. Pérez-de-Heredia, J.A. Luján, S. Zamora, M. Garaulet, Role of DHEA-S on body fat distribution: gender- and depot specific stimulation of adipose tissue lipolysis. Steroids 73, 209–215 (2008)CrossRefPubMed J.J. Hernández-Morante, F. Pérez-de-Heredia, J.A. Luján, S. Zamora, M. Garaulet, Role of DHEA-S on body fat distribution: gender- and depot specific stimulation of adipose tissue lipolysis. Steroids 73, 209–215 (2008)CrossRefPubMed
23.
go back to reference M. Pesaresi, S. Giatti, G. Cavaletti, F. Abbiati, D. Calabrese, R. Lombardi, R. Bianchi, G. Lauria, D. Caruso, L.M. Garcia-Segura, R.C. Melcangi, Sex-dimorphic effects of dehydroepiandrosterone in diabetic neuropathy. Neuroscience 199, 401–409 (2011)CrossRefPubMed M. Pesaresi, S. Giatti, G. Cavaletti, F. Abbiati, D. Calabrese, R. Lombardi, R. Bianchi, G. Lauria, D. Caruso, L.M. Garcia-Segura, R.C. Melcangi, Sex-dimorphic effects of dehydroepiandrosterone in diabetic neuropathy. Neuroscience 199, 401–409 (2011)CrossRefPubMed
24.
go back to reference M.H.V.M. Jacob, D.R. Janner, A. Bello´-Klein, S.F. Llesuy, M.F.M. Ribeiro, Dehydroepiandrosterone modulates antioxidant enzymes and Akt signaling in healthy Wistar rat hearts. J. Steroid Biochem. Mol. Biol. 112, 138–144 (2008)CrossRefPubMed M.H.V.M. Jacob, D.R. Janner, A. Bello´-Klein, S.F. Llesuy, M.F.M. Ribeiro, Dehydroepiandrosterone modulates antioxidant enzymes and Akt signaling in healthy Wistar rat hearts. J. Steroid Biochem. Mol. Biol. 112, 138–144 (2008)CrossRefPubMed
25.
go back to reference M.H.V.M. Jacob, D.R. Janner, M.P. Jahn, L.C.R. Kucharski, A. Belló-Klein, M.F.M. Ribeiro, DHEA effects on myocardial Akt signaling modulation and oxidative stress changes in aged rats. Steroids 74, 1045–1050 (2009)CrossRefPubMed M.H.V.M. Jacob, D.R. Janner, M.P. Jahn, L.C.R. Kucharski, A. Belló-Klein, M.F.M. Ribeiro, DHEA effects on myocardial Akt signaling modulation and oxidative stress changes in aged rats. Steroids 74, 1045–1050 (2009)CrossRefPubMed
26.
go back to reference M.H.V.M. Jacob, D.R. Janner, A.S.R. Araújo, M.P. Jahn, L.C.R. Kucharski, T.B. Moraes, C.S. Dutra Filho, M.F.M. Ribeiro, A. Belló-Klein, Dehydroepiandrosterone improves hepatic antioxidant reserve and stimulates Akt signaling in young and old rats. J. Steroid Biochem. Mol. Biol. 127, 331–336 (2011)CrossRefPubMed M.H.V.M. Jacob, D.R. Janner, A.S.R. Araújo, M.P. Jahn, L.C.R. Kucharski, T.B. Moraes, C.S. Dutra Filho, M.F.M. Ribeiro, A. Belló-Klein, Dehydroepiandrosterone improves hepatic antioxidant reserve and stimulates Akt signaling in young and old rats. J. Steroid Biochem. Mol. Biol. 127, 331–336 (2011)CrossRefPubMed
27.
go back to reference M.P. Jahn, L.F. Gomes, M.H. Jacob, D. da Rocha Janner, A.S. Araújo, A. Belló-Klein, M.F. Ribeiro, L.C. Kucharski, The effect of dehydroepiandrosterone (DHEA) on renal function and metabolism in diabetic rats. Steroids 76, 564–570 (2011)CrossRefPubMed M.P. Jahn, L.F. Gomes, M.H. Jacob, D. da Rocha Janner, A.S. Araújo, A. Belló-Klein, M.F. Ribeiro, L.C. Kucharski, The effect of dehydroepiandrosterone (DHEA) on renal function and metabolism in diabetic rats. Steroids 76, 564–570 (2011)CrossRefPubMed
28.
go back to reference M.C. Castro, F. Francini, J.J. Gagliardino, M.L. Massa, Lipoic acid prevents fructose-induced changes in liver carbohydrate metabolism: role of oxidative stress. Biochim. Biophys. Acta 1840, 1145–1151 (2013)CrossRefPubMed M.C. Castro, F. Francini, J.J. Gagliardino, M.L. Massa, Lipoic acid prevents fructose-induced changes in liver carbohydrate metabolism: role of oxidative stress. Biochim. Biophys. Acta 1840, 1145–1151 (2013)CrossRefPubMed
29.
go back to reference A.G. Ferreira, A.A. da Cunha, F.R. Machado, C.D. Pederzolli, G.R. Dalazen, A.M. de Assis, M.L. Lamers, M.F. dos Santos, C.S. Dutra-Filho, A.T. Wyse, Experimental hyperprolinemia induces mild oxidative stress, metabolic changes and tissue adaptation in rat liver. J. Cell. Biochem. 113, 174–183 (2012)CrossRefPubMed A.G. Ferreira, A.A. da Cunha, F.R. Machado, C.D. Pederzolli, G.R. Dalazen, A.M. de Assis, M.L. Lamers, M.F. dos Santos, C.S. Dutra-Filho, A.T. Wyse, Experimental hyperprolinemia induces mild oxidative stress, metabolic changes and tissue adaptation in rat liver. J. Cell. Biochem. 113, 174–183 (2012)CrossRefPubMed
30.
go back to reference D.R. Janner, M.H.V.M. Jacob, M.P. Jahn, L.C. Kucharski, M.F.M. Ribeiro, Dehydroepiandrosterone effects on Akt signaling modulation in central nervous system of young and aged healthy rats. J. Steroid Biochem. Mol. Biol. 122, 142–148 (2010)CrossRef D.R. Janner, M.H.V.M. Jacob, M.P. Jahn, L.C. Kucharski, M.F.M. Ribeiro, Dehydroepiandrosterone effects on Akt signaling modulation in central nervous system of young and aged healthy rats. J. Steroid Biochem. Mol. Biol. 122, 142–148 (2010)CrossRef
31.
go back to reference M.M. Bradford, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72, 248–254 (1976)CrossRefPubMed M.M. Bradford, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72, 248–254 (1976)CrossRefPubMed
32.
go back to reference L.M. Brown, D.J. Clegg, Central effects of estradiol in the regulation of food intake, body weight, and adiposity. J. Steroid Biochem. Mol. Biol. 122, 65–73 (2010)CrossRefPubMedCentralPubMed L.M. Brown, D.J. Clegg, Central effects of estradiol in the regulation of food intake, body weight, and adiposity. J. Steroid Biochem. Mol. Biol. 122, 65–73 (2010)CrossRefPubMedCentralPubMed
33.
34.
go back to reference K. Esposito, A. Capuano, D. Giugliano, Metabolic syndrome and cancer: holistic or reductionist? Endocrine 45, 362–364 (2014)CrossRefPubMed K. Esposito, A. Capuano, D. Giugliano, Metabolic syndrome and cancer: holistic or reductionist? Endocrine 45, 362–364 (2014)CrossRefPubMed
35.
go back to reference K. Esposito, P. Chiodini, M.I. Maiorino, G. Bellastella, D. Panagiotakos, D. Giugliano, Which diet for prevention of type 2 diabetes? A meta-analysis of prospective studies. Endocrine (2014). doi:10.1007/s12020-014-0264-4 K. Esposito, P. Chiodini, M.I. Maiorino, G. Bellastella, D. Panagiotakos, D. Giugliano, Which diet for prevention of type 2 diabetes? A meta-analysis of prospective studies. Endocrine (2014). doi:10.​1007/​s12020-014-0264-4
36.
go back to reference L. Velázquez-López, E. González-Figueroa, P. Medina-Bravo, I. Pineda-Del Aguila, L. Avila-Jiménez, R. Ramos-Hernández, M. Klunder-Klunder, J. Escobedo de la Peña, Low calorie and carbohydrate diet:to improve the cardiovascular riskindicators in overweightor obese adults with prediabetes. Endocrine 43, 593–602 (2013)CrossRefPubMed L. Velázquez-López, E. González-Figueroa, P. Medina-Bravo, I. Pineda-Del Aguila, L. Avila-Jiménez, R. Ramos-Hernández, M. Klunder-Klunder, J. Escobedo de la Peña, Low calorie and carbohydrate diet:to improve the cardiovascular riskindicators in overweightor obese adults with prediabetes. Endocrine 43, 593–602 (2013)CrossRefPubMed
37.
go back to reference A.L. Hoefel, F. Hansen, P.D. Rosa, A.M. Assis, S.L. Silveira, C.C. Denardin, L. Pettenuzzo, P.R. Augusti, S. Somacal, T. Emanuelli, M.L. Perry, C.M. Wannmacher, The effects of hypercaloric diets on glucose homeostasis in the rat: influence of saturated and monounsaturated dietary lipids. Cell Biochem. Funct. 29, 569–576 (2011)CrossRefPubMed A.L. Hoefel, F. Hansen, P.D. Rosa, A.M. Assis, S.L. Silveira, C.C. Denardin, L. Pettenuzzo, P.R. Augusti, S. Somacal, T. Emanuelli, M.L. Perry, C.M. Wannmacher, The effects of hypercaloric diets on glucose homeostasis in the rat: influence of saturated and monounsaturated dietary lipids. Cell Biochem. Funct. 29, 569–576 (2011)CrossRefPubMed
38.
go back to reference Y. Ono, H. Sorimachi, Calpains: an elaborate proteolytic system. Biochim. Biophys. Acta 1824, 224–236 (2012)CrossRefPubMed Y. Ono, H. Sorimachi, Calpains: an elaborate proteolytic system. Biochim. Biophys. Acta 1824, 224–236 (2012)CrossRefPubMed
39.
go back to reference A.M. Baviera, N.M. Zanon, L.C. Carvalho Navegantes, R.H. Migliorini, I. do Carmo Kettelhut, Pentoxifylline inhibits Ca2+ -dependent and ATP proteasome-dependent proteolysis in skeletal muscle from acutely diabetic rats. Am. J. Physiol. Endocrinol. Metab. 292, E702–E708 (2007)CrossRefPubMed A.M. Baviera, N.M. Zanon, L.C. Carvalho Navegantes, R.H. Migliorini, I. do Carmo Kettelhut, Pentoxifylline inhibits Ca2+ -dependent and ATP proteasome-dependent proteolysis in skeletal muscle from acutely diabetic rats. Am. J. Physiol. Endocrinol. Metab. 292, E702–E708 (2007)CrossRefPubMed
40.
go back to reference C.T. Montague, S. O’Rahilly, Perspectives in diabetes: the perils of portliness causes and consequences of visceral adiposity. Diabetes 49, 883–888 (2000)CrossRefPubMed C.T. Montague, S. O’Rahilly, Perspectives in diabetes: the perils of portliness causes and consequences of visceral adiposity. Diabetes 49, 883–888 (2000)CrossRefPubMed
41.
go back to reference K.F. Petersen, S. Dufour, D. Befroy, M. Lehrke, R.E. Hendler, G.I. Shulman, Reversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hyperglycemia by moderate weight reduction in patients with type 2 diabetes. Diabetes 54, 603–608 (2005)CrossRefPubMedCentralPubMed K.F. Petersen, S. Dufour, D. Befroy, M. Lehrke, R.E. Hendler, G.I. Shulman, Reversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hyperglycemia by moderate weight reduction in patients with type 2 diabetes. Diabetes 54, 603–608 (2005)CrossRefPubMedCentralPubMed
42.
go back to reference S. Galic, J.S. Oakhill, G.R. Steinberg, Adipose tissue as an endocrine organ. Mol. Cell. Endocrinol. 316, 129–139 (2010)CrossRefPubMed S. Galic, J.S. Oakhill, G.R. Steinberg, Adipose tissue as an endocrine organ. Mol. Cell. Endocrinol. 316, 129–139 (2010)CrossRefPubMed
43.
go back to reference S.L. Doyle, C.L. Donohoe, J. Lysaght, J.V. Reynolds, Visceral obesity, metabolic syndrome, insulin resistance and cancer. Proc. Nutr. Soc. 71, 181–189 (2012)CrossRefPubMed S.L. Doyle, C.L. Donohoe, J. Lysaght, J.V. Reynolds, Visceral obesity, metabolic syndrome, insulin resistance and cancer. Proc. Nutr. Soc. 71, 181–189 (2012)CrossRefPubMed
44.
go back to reference B.L. Wajchenberg, Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr. Rev. 21, 697–738 (2000)CrossRefPubMed B.L. Wajchenberg, Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr. Rev. 21, 697–738 (2000)CrossRefPubMed
46.
47.
go back to reference R. Lomonaco, N.E. Sunny, F. Bril, K. Cusi, Nonalcoholic fatty liver disease: current issues and novel treatment approaches. Drugs 73, 1–14 (2013)CrossRefPubMed R. Lomonaco, N.E. Sunny, F. Bril, K. Cusi, Nonalcoholic fatty liver disease: current issues and novel treatment approaches. Drugs 73, 1–14 (2013)CrossRefPubMed
48.
go back to reference D. Constantin-Teodosiu, Regulation of muscle pyruvate dehydrogenase complex in insulin resistance: effects of exercise and dichloroacetate. Diabetes Metab. J. 37, 301–314 (2013)CrossRefPubMedCentralPubMed D. Constantin-Teodosiu, Regulation of muscle pyruvate dehydrogenase complex in insulin resistance: effects of exercise and dichloroacetate. Diabetes Metab. J. 37, 301–314 (2013)CrossRefPubMedCentralPubMed
49.
go back to reference J.O. Lee, S.K. Lee, J.H. Jung, J.H. Kim, G.Y. You, S.J. Kim, S.H. Park, K.O. Uhm, H.S. Kim, Metformin induces Rab4 through AMPK and modulates GLUT4 translocation in skeletal muscle cells. J. Cell. Physiol. 226, 974–981 (2011)CrossRefPubMed J.O. Lee, S.K. Lee, J.H. Jung, J.H. Kim, G.Y. You, S.J. Kim, S.H. Park, K.O. Uhm, H.S. Kim, Metformin induces Rab4 through AMPK and modulates GLUT4 translocation in skeletal muscle cells. J. Cell. Physiol. 226, 974–981 (2011)CrossRefPubMed
50.
go back to reference A. Nadal-Casellas, A.M. Proenza, I. Lladó, M. Gianotti, Sex-dependent differences in rat hepatic lipid accumulation and insulin sensitivity in response to diet-induced obesity. Biochem. Cell Biol. 90, 164–172 (2012)CrossRefPubMed A. Nadal-Casellas, A.M. Proenza, I. Lladó, M. Gianotti, Sex-dependent differences in rat hepatic lipid accumulation and insulin sensitivity in response to diet-induced obesity. Biochem. Cell Biol. 90, 164–172 (2012)CrossRefPubMed
51.
go back to reference S. Kumagai, A. Holmäng, P. Björntorp, The effects of oestrogen and progesterone on insulin sensitivity in female rats. Acta Physiol. Scand. 149, 91–97 (1993)CrossRefPubMed S. Kumagai, A. Holmäng, P. Björntorp, The effects of oestrogen and progesterone on insulin sensitivity in female rats. Acta Physiol. Scand. 149, 91–97 (1993)CrossRefPubMed
52.
go back to reference T. Jelenik, M. Roden, How estrogens prevent from lipid-induced insulin resistance. Endocrinology 154, 989–992 (2013)CrossRefPubMed T. Jelenik, M. Roden, How estrogens prevent from lipid-induced insulin resistance. Endocrinology 154, 989–992 (2013)CrossRefPubMed
53.
go back to reference J.S. Mayes, G.H. Watson, Direct effects of sex steroid hormones on adipose tissues and obesity. Obes. Rev. 5, 197–216 (2004)CrossRefPubMed J.S. Mayes, G.H. Watson, Direct effects of sex steroid hormones on adipose tissues and obesity. Obes. Rev. 5, 197–216 (2004)CrossRefPubMed
54.
go back to reference S. Stöppeler, D. Palmes, M. Fehr, J.P. Hölzen, A. Zibert, R. Siaj, H.H. Schmidt, H.U. Spiegel, R. Bahde, Gender and strainspecific differences inthe development of steatosis in rats. Lab. Anim. 47, 43–52 (2013)CrossRefPubMed S. Stöppeler, D. Palmes, M. Fehr, J.P. Hölzen, A. Zibert, R. Siaj, H.H. Schmidt, H.U. Spiegel, R. Bahde, Gender and strainspecific differences inthe development of steatosis in rats. Lab. Anim. 47, 43–52 (2013)CrossRefPubMed
Metadata
Title
Sex-related differences in the effects of high-fat diets on DHEA-treated rats
Authors
Ana Lúcia Cecconello
Marcia Trapp
Ana Lúcia Hoefel
Cláudia Vieira Marques
Bruno Dutra Arbo
Gabriela Osterkamp
Luiz Carlos Rios Kucharski
Maria Flávia Marques Ribeiro
Publication date
01-04-2015
Publisher
Springer US
Published in
Endocrine / Issue 3/2015
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-014-0396-6

Other articles of this Issue 3/2015

Endocrine 3/2015 Go to the issue