Skip to main content
Erschienen in: Endocrine 3/2012

01.12.2012 | Original Article

The effect of chronic immobilization stress on leptin signaling in the ovariectomized (OVX) rat

verfasst von: Darwin O. Larco, Danette F. Cruthirds, Michael J. Weiser, Robert J. Handa, T. John Wu

Erschienen in: Endocrine | Ausgabe 3/2012

Einloggen, um Zugang zu erhalten

Abstract

Previous studies have shown that both 17β-estradiol (E2) treatment and chronic stress may attenuate post-OVX weight gain in the female rat. However, the interaction between E2 and stress is unclear. This study examined the effect of E2 treatment and chronic immobilization stress on body weight. Adult OVX Sprague–Dawley rats were randomly assigned to one of four treatment groups in a 2X2 factorial design examining hormone treatment [vehicle (VEH) or E2, sc] and stress (no stress vs stress 60 min/day for 22 days). After 22 days, E2 significantly inhibited weight gain and food intake in OVX rats. In contrast, chronic stress reduced body weight only in control OVX animals but did not affect food intake. E2 reduced circulating leptin levels in non-stressed animals, but not in animals subjected to chronic immobilization. Western blot analysis indicated that E2 treatment increased leptin receptor (Ob-Rb) expression in the medial basal hypothalamus (MBH); however, this treatment also increased suppressor of cytokine signaling 3 (SOCS3), which is an inhibitor of leptin signaling. Chronic immobilization stress blunted the E2-induced increase in Ob-Rb and SOCS3 levels. These results suggest that chronic stress counteracts E2 effects on leptin signaling in the MBH without altering body weight.
Literatur
1.
Zurück zum Zitat T.C. Adam, E.S. Epel, Stress, eating and the reward system. Physiol. Behav. 91, 449–458 (2007)PubMedCrossRef T.C. Adam, E.S. Epel, Stress, eating and the reward system. Physiol. Behav. 91, 449–458 (2007)PubMedCrossRef
2.
Zurück zum Zitat D.A. Ainslie, M.J. Morris, G. Wittert, H. Turnbull, J. Proietto, A.W. Thorburn, Estrogen deficiency causes central leptin insensitivity and increased hypothalamic neuropeptide Y. Int. J. Obes. Relat. Metab. Disord. 25, 1680–1688 (2001)PubMedCrossRef D.A. Ainslie, M.J. Morris, G. Wittert, H. Turnbull, J. Proietto, A.W. Thorburn, Estrogen deficiency causes central leptin insensitivity and increased hypothalamic neuropeptide Y. Int. J. Obes. Relat. Metab. Disord. 25, 1680–1688 (2001)PubMedCrossRef
3.
Zurück zum Zitat A.S. Banks, S.M. Davis, S.H. Bates, M.G. Myers Jr, Activation of downstream signals by the long form of the leptin receptor. J. Biol. Chem. 275, 14563–14572 (2000)PubMedCrossRef A.S. Banks, S.M. Davis, S.H. Bates, M.G. Myers Jr, Activation of downstream signals by the long form of the leptin receptor. J. Biol. Chem. 275, 14563–14572 (2000)PubMedCrossRef
4.
Zurück zum Zitat W.A. Banks, The blood–brain barrier as a cause of obesity. Curr. Pharm. Des. 14, 1606–1614 (2008)PubMedCrossRef W.A. Banks, The blood–brain barrier as a cause of obesity. Curr. Pharm. Des. 14, 1606–1614 (2008)PubMedCrossRef
5.
Zurück zum Zitat C. Bjorbaek, K. El-Haschimi, J.D. Frantz, J.S. Flier, The role of SOCS-3 in leptin signaling and leptin resistance. J. Biol. Chem. 274, 30059–30065 (1999)PubMedCrossRef C. Bjorbaek, K. El-Haschimi, J.D. Frantz, J.S. Flier, The role of SOCS-3 in leptin signaling and leptin resistance. J. Biol. Chem. 274, 30059–30065 (1999)PubMedCrossRef
6.
Zurück zum Zitat C. Bjorbaek, J.K. Elmquist, J.D. Frantz, S.E. Shoelson, J.S. Flier, Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol. Cell 1, 619–625 (1998)PubMedCrossRef C. Bjorbaek, J.K. Elmquist, J.D. Frantz, S.E. Shoelson, J.S. Flier, Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol. Cell 1, 619–625 (1998)PubMedCrossRef
7.
Zurück zum Zitat A.L. Boutillier, D. Monnier, D. Lorang, J.R. Lundblad, J.L. Roberts, J.P. Loeffler, Corticotropin-releasing hormone stimulates proopiomelanocortin transcription by cFos-dependent and -independent pathways: characterization of an AP1 site in exon 1. Mol. Endocrinol. 9, 745–755 (1995)PubMedCrossRef A.L. Boutillier, D. Monnier, D. Lorang, J.R. Lundblad, J.L. Roberts, J.P. Loeffler, Corticotropin-releasing hormone stimulates proopiomelanocortin transcription by cFos-dependent and -independent pathways: characterization of an AP1 site in exon 1. Mol. Endocrinol. 9, 745–755 (1995)PubMedCrossRef
8.
Zurück zum Zitat R.E. Bowman, D. Ferguson, V.N. Luine, Effects of chronic restraint stress and estradiol on open field activity, spatial memory, and monoaminergic neurotransmitters in ovariectomized rats. Neuroscience 113, 401–410 (2002)PubMedCrossRef R.E. Bowman, D. Ferguson, V.N. Luine, Effects of chronic restraint stress and estradiol on open field activity, spatial memory, and monoaminergic neurotransmitters in ovariectomized rats. Neuroscience 113, 401–410 (2002)PubMedCrossRef
9.
Zurück zum Zitat S. Chakraborty, A. Sachdev, S.R. Salton, T.R. Chakraborty, Stereological analysis of estrogen receptor expression in the hypothalamic arcuate nucleus of ob/ob and agouti mice. Brain Res. 1217, 86–95 (2008)PubMedCrossRef S. Chakraborty, A. Sachdev, S.R. Salton, T.R. Chakraborty, Stereological analysis of estrogen receptor expression in the hypothalamic arcuate nucleus of ob/ob and agouti mice. Brain Res. 1217, 86–95 (2008)PubMedCrossRef
10.
Zurück zum Zitat F. Chigr, F. Rachidi, S. Segura, S. Mahaut, C. Tardivel, A. Jean, M. Najimi, E. Moyse, Neurogenesis inhibition in the dorsal vagal complex by chronic immobilization stress in the adult rat. Neuroscience 158, 524–536 (2009)PubMedCrossRef F. Chigr, F. Rachidi, S. Segura, S. Mahaut, C. Tardivel, A. Jean, M. Najimi, E. Moyse, Neurogenesis inhibition in the dorsal vagal complex by chronic immobilization stress in the adult rat. Neuroscience 158, 524–536 (2009)PubMedCrossRef
11.
Zurück zum Zitat S.C. Chua Jr, W.K. Chung, X.S. Wu-Peng, Y. Zhang, S.M. Liu, L. Tartaglia, R.L. Leibel, Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science 271, 994–996 (1996)PubMedCrossRef S.C. Chua Jr, W.K. Chung, X.S. Wu-Peng, Y. Zhang, S.M. Liu, L. Tartaglia, R.L. Leibel, Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science 271, 994–996 (1996)PubMedCrossRef
12.
Zurück zum Zitat P.J. Crack, T.J. Wu, P.M. Cummins, E.S. Ferro, J.W. Tullai, M.J. Glucksman, J.L. Roberts, The association of metalloendopeptidase EC 3.4.24.15 at the extracellular surface of the AtT-20 cell plasma membrane. Brain Res. 835, 113–124 (1999)PubMedCrossRef P.J. Crack, T.J. Wu, P.M. Cummins, E.S. Ferro, J.W. Tullai, M.J. Glucksman, J.L. Roberts, The association of metalloendopeptidase EC 3.4.24.15 at the extracellular surface of the AtT-20 cell plasma membrane. Brain Res. 835, 113–124 (1999)PubMedCrossRef
13.
Zurück zum Zitat K. Dechering, C. Boersma, S. Mosselman, Estrogen receptors alpha and beta: two receptors of a kind? Curr. Med. Chem. 7, 561–576 (2000)PubMedCrossRef K. Dechering, C. Boersma, S. Mosselman, Estrogen receptors alpha and beta: two receptors of a kind? Curr. Med. Chem. 7, 561–576 (2000)PubMedCrossRef
14.
Zurück zum Zitat E. Enmark, J.A. Gustafsson, Oestrogen receptors—an overview. J. Intern. Med. 246, 133–138 (1999)PubMedCrossRef E. Enmark, J.A. Gustafsson, Oestrogen receptors—an overview. J. Intern. Med. 246, 133–138 (1999)PubMedCrossRef
15.
Zurück zum Zitat M.M. Faraday, Rat sex and strain differences in responses to stress. Physiol. Behav. 75, 507–522 (2002)PubMedCrossRef M.M. Faraday, Rat sex and strain differences in responses to stress. Physiol. Behav. 75, 507–522 (2002)PubMedCrossRef
16.
Zurück zum Zitat M.M. Faraday, K.H. Blakeman, N.E. Grunberg, Strain and sex alter effects of stress and nicotine on feeding, body weight, and HPA axis hormones. Pharmacol. Biochem. Behav. 80, 577–589 (2005)PubMedCrossRef M.M. Faraday, K.H. Blakeman, N.E. Grunberg, Strain and sex alter effects of stress and nicotine on feeding, body weight, and HPA axis hormones. Pharmacol. Biochem. Behav. 80, 577–589 (2005)PubMedCrossRef
17.
Zurück zum Zitat M. Gambacciani, M. Ciaponi, B. Cappagli, L. De Simone, R. Orlandi, A.R. Genazzani, Prospective evaluation of body weight and body fat distribution in early postmenopausal women with and without hormonal replacement therapy. Maturitas 39, 125–132 (2001)PubMedCrossRef M. Gambacciani, M. Ciaponi, B. Cappagli, L. De Simone, R. Orlandi, A.R. Genazzani, Prospective evaluation of body weight and body fat distribution in early postmenopausal women with and without hormonal replacement therapy. Maturitas 39, 125–132 (2001)PubMedCrossRef
18.
Zurück zum Zitat Q. Gao, T.L. Horvath, Neurobiology of feeding and energy expenditure. Annu. Rev. Neurosci. 30, 367–398 (2007)PubMedCrossRef Q. Gao, T.L. Horvath, Neurobiology of feeding and energy expenditure. Annu. Rev. Neurosci. 30, 367–398 (2007)PubMedCrossRef
19.
Zurück zum Zitat Q. Gao, T.L. Horvath, Cross-talk between estrogen and leptin signaling in the hypothalamus. Am. J. Physiol. Endocrinol. Metab. 294, E817–E826 (2008)PubMedCrossRef Q. Gao, T.L. Horvath, Cross-talk between estrogen and leptin signaling in the hypothalamus. Am. J. Physiol. Endocrinol. Metab. 294, E817–E826 (2008)PubMedCrossRef
20.
Zurück zum Zitat Q. Gao, G. Mezei, Y. Nie, Y. Rao, C.S. Choi, I. Bechmann, C. Leranth, D. Toran-Allerand, C.A. Priest, J.L. Roberts, X.B. Gao, C. Mobbs, G.I. Shulman, S. Diano, T.L. Horvath, Anorectic estrogen mimics leptin’s effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals. Nat. Med. 13, 89–94 (2007)PubMedCrossRef Q. Gao, G. Mezei, Y. Nie, Y. Rao, C.S. Choi, I. Bechmann, C. Leranth, D. Toran-Allerand, C.A. Priest, J.L. Roberts, X.B. Gao, C. Mobbs, G.I. Shulman, S. Diano, T.L. Horvath, Anorectic estrogen mimics leptin’s effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals. Nat. Med. 13, 89–94 (2007)PubMedCrossRef
21.
Zurück zum Zitat S. Gao, K.P. Kinzig, S. Aja, K.A. Scott, W. Keung, S. Kelly, K. Strynadka, S. Chohnan, W.W. Smith, K.L. Tamashiro, E.E. Ladenheim, G.V. Ronnett, Y. Tu, M.J. Birnbaum, G.D. Lopaschuk, T.H. Moran, Leptin activates hypothalamic acetyl-CoA carboxylase to inhibit food intake. Proc Natl Acad Sci USA 104, 17358–17363 (2007)PubMedCrossRef S. Gao, K.P. Kinzig, S. Aja, K.A. Scott, W. Keung, S. Kelly, K. Strynadka, S. Chohnan, W.W. Smith, K.L. Tamashiro, E.E. Ladenheim, G.V. Ronnett, Y. Tu, M.J. Birnbaum, G.D. Lopaschuk, T.H. Moran, Leptin activates hypothalamic acetyl-CoA carboxylase to inhibit food intake. Proc Natl Acad Sci USA 104, 17358–17363 (2007)PubMedCrossRef
22.
Zurück zum Zitat J. Haarbo, U. Marslew, A. Gotfredsen, C. Christiansen, Postmenopausal hormone replacement therapy prevents central distribution of body fat after menopause. Metabolism 40, 1323–1326 (1991)PubMedCrossRef J. Haarbo, U. Marslew, A. Gotfredsen, C. Christiansen, Postmenopausal hormone replacement therapy prevents central distribution of body fat after menopause. Metabolism 40, 1323–1326 (1991)PubMedCrossRef
23.
Zurück zum Zitat R.J. Handa, D.L. Reid, J.A. Resko, Androgen receptors in brain and pituitary of female rats: cyclic changes and comparisons with the male. Biol. Reprod. 34, 293–303 (1986)PubMedCrossRef R.J. Handa, D.L. Reid, J.A. Resko, Androgen receptors in brain and pituitary of female rats: cyclic changes and comparisons with the male. Biol. Reprod. 34, 293–303 (1986)PubMedCrossRef
24.
Zurück zum Zitat M. Hotta, T. Shibasaki, N. Yamauchi, H. Ohno, R. Benoit, N. Ling, H. Demura, The effects of chronic central administration of corticotropin-releasing factor on food intake, body weight, and hypothalamic–pituitary–adrenocortical hormones. Life Sci. 48, 1483–1491 (1991)PubMedCrossRef M. Hotta, T. Shibasaki, N. Yamauchi, H. Ohno, R. Benoit, N. Ling, H. Demura, The effects of chronic central administration of corticotropin-releasing factor on food intake, body weight, and hypothalamic–pituitary–adrenocortical hormones. Life Sci. 48, 1483–1491 (1991)PubMedCrossRef
25.
Zurück zum Zitat J.W. Jahng, N.Y. Kim, V. Ryu, S.B. Yoo, B.T. Kim, D.W. Kang, J.H. Lee, Dexamethasone reduces food intake, weight gain and the hypothalamic 5-HT concentration and increases plasma leptin in rats. Eur. J. Pharmacol. 581, 64–70 (2008)PubMedCrossRef J.W. Jahng, N.Y. Kim, V. Ryu, S.B. Yoo, B.T. Kim, D.W. Kang, J.H. Lee, Dexamethasone reduces food intake, weight gain and the hypothalamic 5-HT concentration and increases plasma leptin in rats. Eur. J. Pharmacol. 581, 64–70 (2008)PubMedCrossRef
26.
Zurück zum Zitat H. Jin, N.M. Lanouette, S. Mudaliar, R. Henry, D.P. Folsom, S. Khandrika, D.K. Glorioso, D.V. Jeste, Association of posttraumatic stress disorder with increased prevalence of metabolic syndrome. J. Clin. Psychopharmacol. 29, 210–215 (2009)PubMedCrossRef H. Jin, N.M. Lanouette, S. Mudaliar, R. Henry, D.P. Folsom, S. Khandrika, D.K. Glorioso, D.V. Jeste, Association of posttraumatic stress disorder with increased prevalence of metabolic syndrome. J. Clin. Psychopharmacol. 29, 210–215 (2009)PubMedCrossRef
27.
Zurück zum Zitat J.M. Joyner, L.J. Hutley, D.P. Cameron, Estrogen receptors in human preadipocytes. Endocrine 15, 225–230 (2001)PubMedCrossRef J.M. Joyner, L.J. Hutley, D.P. Cameron, Estrogen receptors in human preadipocytes. Endocrine 15, 225–230 (2001)PubMedCrossRef
28.
Zurück zum Zitat P.A. Lapchak, F. Hefti, BDNF and NGF treatment in lesioned rats: effects on cholinergic function and weight gain. NeuroReport 3, 405–408 (1992)PubMedCrossRef P.A. Lapchak, F. Hefti, BDNF and NGF treatment in lesioned rats: effects on cholinergic function and weight gain. NeuroReport 3, 405–408 (1992)PubMedCrossRef
29.
Zurück zum Zitat D.W. Leaman, S. Leung, X. Li, G.R. Stark, Regulation of STAT-dependent pathways by growth factors and cytokines. FASEB J. 10, 1578–1588 (1996)PubMed D.W. Leaman, S. Leung, X. Li, G.R. Stark, Regulation of STAT-dependent pathways by growth factors and cytokines. FASEB J. 10, 1578–1588 (1996)PubMed
30.
Zurück zum Zitat T.D. Lund, L.R. Hinds, R.J. Handa, The androgen 5alpha-dihydrotestosterone and its metabolite 5alpha-androstan-3beta, 17beta-diol inhibit the hypothalamo–pituitary–adrenal response to stress by acting through estrogen receptor beta-expressing neurons in the hypothalamus. J. Neurosci. 26, 1448–1456 (2006)PubMedCrossRef T.D. Lund, L.R. Hinds, R.J. Handa, The androgen 5alpha-dihydrotestosterone and its metabolite 5alpha-androstan-3beta, 17beta-diol inhibit the hypothalamo–pituitary–adrenal response to stress by acting through estrogen receptor beta-expressing neurons in the hypothalamus. J. Neurosci. 26, 1448–1456 (2006)PubMedCrossRef
31.
Zurück zum Zitat F. Machinal-Quelin, M.N. Dieudonne, R. Pecquery, M.C. Leneveu, Y. Giudicelli, Direct in vitro effects of androgens and estrogens on ob gene expression and leptin secretion in human adipose tissue. Endocrine 18, 179–184 (2002)PubMedCrossRef F. Machinal-Quelin, M.N. Dieudonne, R. Pecquery, M.C. Leneveu, Y. Giudicelli, Direct in vitro effects of androgens and estrogens on ob gene expression and leptin secretion in human adipose tissue. Endocrine 18, 179–184 (2002)PubMedCrossRef
32.
Zurück zum Zitat M. Maffei, J. Halaas, E. Ravussin, R.E. Pratley, G.H. Lee, Y. Zhang, H. Fei, S. Kim, R. Lallone, S. Ranganathan et al., Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat. Med. 1, 1155–1161 (1995)PubMedCrossRef M. Maffei, J. Halaas, E. Ravussin, R.E. Pratley, G.H. Lee, Y. Zhang, H. Fei, S. Kim, R. Lallone, S. Ranganathan et al., Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat. Med. 1, 1155–1161 (1995)PubMedCrossRef
33.
Zurück zum Zitat S. Makino, K. Asaba, M. Nishiyama, K. Hashimoto, Decreased type 2 corticotropin-releasing hormone receptor mRNA expression in the ventromedial hypothalamus during repeated immobilization stress. Neuroendocrinology 70, 160–167 (1999)PubMedCrossRef S. Makino, K. Asaba, M. Nishiyama, K. Hashimoto, Decreased type 2 corticotropin-releasing hormone receptor mRNA expression in the ventromedial hypothalamus during repeated immobilization stress. Neuroendocrinology 70, 160–167 (1999)PubMedCrossRef
34.
Zurück zum Zitat G. Mastorakos, E. Zapanti, The hypothalamic–pituitary–adrenal axis in the neuroendocrine regulation of food intake and obesity: the role of corticotropin releasing hormone. Nutr. Neurosci. 7, 271–280 (2004)PubMedCrossRef G. Mastorakos, E. Zapanti, The hypothalamic–pituitary–adrenal axis in the neuroendocrine regulation of food intake and obesity: the role of corticotropin releasing hormone. Nutr. Neurosci. 7, 271–280 (2004)PubMedCrossRef
35.
Zurück zum Zitat K.M. McCormick, K.L. Burns, C.M. Piccone, L.E. Gosselin, G.A. Brazeau, Effects of ovariectomy and estrogen on skeletal muscle function in growing rats. J. Muscle Res. Cell Motil. 25, 21–27 (2004)PubMedCrossRef K.M. McCormick, K.L. Burns, C.M. Piccone, L.E. Gosselin, G.A. Brazeau, Effects of ovariectomy and estrogen on skeletal muscle function in growing rats. J. Muscle Res. Cell Motil. 25, 21–27 (2004)PubMedCrossRef
36.
Zurück zum Zitat R. Meli, M. Pacilio, G.M. Raso, E. Esposito, A. Coppola, A. Nasti, C. Di Carlo, C. Nappi, R. Di Carlo, Estrogen and raloxifene modulate leptin and its receptor in hypothalamus and adipose tissue from ovariectomized rats. Endocrinology 145, 3115–3121 (2004)PubMedCrossRef R. Meli, M. Pacilio, G.M. Raso, E. Esposito, A. Coppola, A. Nasti, C. Di Carlo, C. Nappi, R. Di Carlo, Estrogen and raloxifene modulate leptin and its receptor in hypothalamus and adipose tissue from ovariectomized rats. Endocrinology 145, 3115–3121 (2004)PubMedCrossRef
37.
Zurück zum Zitat I. Morimoto, S. Yamamoto, K. Kai, T. Fujihira, E. Morita, S. Eto, Centrally administered murine–leptin stimulates the hypothalamus–pituitary–adrenal axis through arginine–vasopressin. Neuroendocrinology 71, 366–374 (2000)PubMedCrossRef I. Morimoto, S. Yamamoto, K. Kai, T. Fujihira, E. Morita, S. Eto, Centrally administered murine–leptin stimulates the hypothalamus–pituitary–adrenal axis through arginine–vasopressin. Neuroendocrinology 71, 366–374 (2000)PubMedCrossRef
38.
Zurück zum Zitat H. Munzberg, J.S. Flier, C. Bjorbaek, Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology 145, 4880–4889 (2004)PubMedCrossRef H. Munzberg, J.S. Flier, C. Bjorbaek, Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology 145, 4880–4889 (2004)PubMedCrossRef
39.
Zurück zum Zitat H. Munzberg, L. Huo, E.A. Nillni, A.N. Hollenberg, C. Bjorbaek, Role of signal transducer and activator of transcription 3 in regulation of hypothalamic proopiomelanocortin gene expression by leptin. Endocrinology 144, 2121–2131 (2003)PubMedCrossRef H. Munzberg, L. Huo, E.A. Nillni, A.N. Hollenberg, C. Bjorbaek, Role of signal transducer and activator of transcription 3 in regulation of hypothalamic proopiomelanocortin gene expression by leptin. Endocrinology 144, 2121–2131 (2003)PubMedCrossRef
40.
Zurück zum Zitat G. Naert, G. Ixart, T. Maurice, L. Tapia-Arancibia, L. Givalois, Brain-derived neurotrophic factor and hypothalamic–pituitary–adrenal axis adaptation processes in a depressive-like state induced by chronic restraint stress. Mol. Cell. Neurosci. 46, 55–66 (2011)PubMedCrossRef G. Naert, G. Ixart, T. Maurice, L. Tapia-Arancibia, L. Givalois, Brain-derived neurotrophic factor and hypothalamic–pituitary–adrenal axis adaptation processes in a depressive-like state induced by chronic restraint stress. Mol. Cell. Neurosci. 46, 55–66 (2011)PubMedCrossRef
41.
Zurück zum Zitat G. Naert, G. Ixart, L. Tapia-Arancibia, L. Givalois, Continuous i.c.v. infusion of brain-derived neurotrophic factor modifies hypothalamic–pituitary–adrenal axis activity, locomotor activity and body temperature rhythms in adult male rats. Neuroscience 139, 779–789 (2006)PubMedCrossRef G. Naert, G. Ixart, L. Tapia-Arancibia, L. Givalois, Continuous i.c.v. infusion of brain-derived neurotrophic factor modifies hypothalamic–pituitary–adrenal axis activity, locomotor activity and body temperature rhythms in adult male rats. Neuroscience 139, 779–789 (2006)PubMedCrossRef
42.
Zurück zum Zitat C. Ohlsson, N. Hellberg, P. Parini, O. Vidal, Y.M. Bohlooly, M. Rudling, M.K. Lindberg, M. Warner, B. Angelin, J.A. Gustafsson, Obesity and disturbed lipoprotein profile in estrogen receptor-alpha-deficient male mice. Biochem. Biophys. Res. Commun. 278, 640–645 (2000)PubMedCrossRef C. Ohlsson, N. Hellberg, P. Parini, O. Vidal, Y.M. Bohlooly, M. Rudling, M.K. Lindberg, M. Warner, B. Angelin, J.A. Gustafsson, Obesity and disturbed lipoprotein profile in estrogen receptor-alpha-deficient male mice. Biochem. Biophys. Res. Commun. 278, 640–645 (2000)PubMedCrossRef
43.
Zurück zum Zitat K. Pettersson, J.A. Gustafsson, Role of estrogen receptor beta in estrogen action. Annu. Rev. Physiol. 63, 165–192 (2001)PubMedCrossRef K. Pettersson, J.A. Gustafsson, Role of estrogen receptor beta in estrogen action. Annu. Rev. Physiol. 63, 165–192 (2001)PubMedCrossRef
44.
Zurück zum Zitat J. Piermaria, G. Console, M. Perello, G. Moreno, R.C. Gaillard, E. Spinedi, Impact of estradiol on parametrial adipose tissue function: evidence for establishment of a new set point of leptin sensitivity in control of energy metabolism in female rat. Endocrine 20, 239–245 (2003)PubMedCrossRef J. Piermaria, G. Console, M. Perello, G. Moreno, R.C. Gaillard, E. Spinedi, Impact of estradiol on parametrial adipose tissue function: evidence for establishment of a new set point of leptin sensitivity in control of energy metabolism in female rat. Endocrine 20, 239–245 (2003)PubMedCrossRef
45.
Zurück zum Zitat A. Pighon, J. Gutkowska, M. Jankowski, R. Rabasa-Lhoret, J.M. Lavoie, Exercise training in ovariectomized rats stimulates estrogenic-like effects on expression of genes involved in lipid accumulation and subclinical inflammation in liver. Metabolism 60(5), 629–639 (2011)PubMedCrossRef A. Pighon, J. Gutkowska, M. Jankowski, R. Rabasa-Lhoret, J.M. Lavoie, Exercise training in ovariectomized rats stimulates estrogenic-like effects on expression of genes involved in lipid accumulation and subclinical inflammation in liver. Metabolism 60(5), 629–639 (2011)PubMedCrossRef
46.
Zurück zum Zitat C.A. Priest, J.L. Roberts, Estrogen and tamoxifen differentially regulate beta-endorphin and cFos expression and neuronal colocalization in the arcuate nucleus of the rat. Neuroendocrinology 72, 293–305 (2000)PubMedCrossRef C.A. Priest, J.L. Roberts, Estrogen and tamoxifen differentially regulate beta-endorphin and cFos expression and neuronal colocalization in the arcuate nucleus of the rat. Neuroendocrinology 72, 293–305 (2000)PubMedCrossRef
47.
Zurück zum Zitat S. Retana-Marquez, H. Bonilla-Jaime, G. Vazquez-Palacios, E. Dominguez-Salazar, R. Martinez-Garcia, J. Velazquez-Moctezuma, Body weight gain and diurnal differences of corticosterone changes in response to acute and chronic stress in rats. Psychoneuroendocrinology 28, 207–227 (2003)PubMedCrossRef S. Retana-Marquez, H. Bonilla-Jaime, G. Vazquez-Palacios, E. Dominguez-Salazar, R. Martinez-Garcia, J. Velazquez-Moctezuma, Body weight gain and diurnal differences of corticosterone changes in response to acute and chronic stress in rats. Psychoneuroendocrinology 28, 207–227 (2003)PubMedCrossRef
48.
Zurück zum Zitat D.M. Roesch, Effects of selective estrogen receptor agonists on food intake and body weight gain in rats. Physiol. Behav. 87, 39–44 (2006)PubMedCrossRef D.M. Roesch, Effects of selective estrogen receptor agonists on food intake and body weight gain in rats. Physiol. Behav. 87, 39–44 (2006)PubMedCrossRef
49.
Zurück zum Zitat C.E. Roselli, S.A. Klosterman, T.A. Fasasi, Sex differences in androgen responsiveness in the rat brain: regional differences in the induction of aromatase activity. Neuroendocrinology 64, 139–145 (1996)PubMedCrossRef C.E. Roselli, S.A. Klosterman, T.A. Fasasi, Sex differences in androgen responsiveness in the rat brain: regional differences in the induction of aromatase activity. Neuroendocrinology 64, 139–145 (1996)PubMedCrossRef
50.
Zurück zum Zitat C. Schindler, J.E. Darnell Jr, Transcriptional responses to polypeptide ligands: the JAK-STAT pathway. Annu. Rev. Biochem. 64, 621–651 (1995)PubMedCrossRef C. Schindler, J.E. Darnell Jr, Transcriptional responses to polypeptide ligands: the JAK-STAT pathway. Annu. Rev. Biochem. 64, 621–651 (1995)PubMedCrossRef
51.
Zurück zum Zitat Y. Shaham, K. Alvares, S.M. Nespor, N.E. Grunberg, Effect of stress on oral morphine and fentanyl self-administration in rats. Pharmacol. Biochem. Behav. 41, 615–619 (1992)PubMedCrossRef Y. Shaham, K. Alvares, S.M. Nespor, N.E. Grunberg, Effect of stress on oral morphine and fentanyl self-administration in rats. Pharmacol. Biochem. Behav. 41, 615–619 (1992)PubMedCrossRef
52.
Zurück zum Zitat F.J. Steyn, G.M. Anderson, D.R. Grattan, Hormonal regulation of suppressors of cytokine signaling (SOCS) messenger ribonucleic acid in the arcuate nucleus during late pregnancy. Endocrinology 149, 3206–3214 (2008)PubMedCrossRef F.J. Steyn, G.M. Anderson, D.R. Grattan, Hormonal regulation of suppressors of cytokine signaling (SOCS) messenger ribonucleic acid in the arcuate nucleus during late pregnancy. Endocrinology 149, 3206–3214 (2008)PubMedCrossRef
53.
Zurück zum Zitat I.N. Wegorzewska, K. Walters, M.J. Weiser, D.F. Cruthirds, E. Ewell, D.O. Larco, R.J. Handa, T.J. Wu, Postovariectomy weight gain in female rats is reversed by estrogen receptor alpha agonist, propylpyrazoletriol. Am. J. Obstet. Gynecol. 199, 67 e61–67 e65 (2008)CrossRef I.N. Wegorzewska, K. Walters, M.J. Weiser, D.F. Cruthirds, E. Ewell, D.O. Larco, R.J. Handa, T.J. Wu, Postovariectomy weight gain in female rats is reversed by estrogen receptor alpha agonist, propylpyrazoletriol. Am. J. Obstet. Gynecol. 199, 67 e61–67 e65 (2008)CrossRef
54.
Zurück zum Zitat M.J. Weiser, T.J. Wu, R.J. Handa, Estrogen receptor-beta agonist diarylpropionitrile: biological activities of R- and S-enantiomers on behavior and hormonal response to stress. Endocrinology 150, 1817–1825 (2009)PubMedCrossRef M.J. Weiser, T.J. Wu, R.J. Handa, Estrogen receptor-beta agonist diarylpropionitrile: biological activities of R- and S-enantiomers on behavior and hormonal response to stress. Endocrinology 150, 1817–1825 (2009)PubMedCrossRef
56.
Zurück zum Zitat H. Yuksel, A.R. Odabasi, S. Demircan, K. Koseoglu, K. Kizilkaya, E. Onur, Effects of postmenopausal hormone replacement therapy on body fat composition. Gynecol. Endocrinol. 23, 99–104 (2007)PubMedCrossRef H. Yuksel, A.R. Odabasi, S. Demircan, K. Koseoglu, K. Kizilkaya, E. Onur, Effects of postmenopausal hormone replacement therapy on body fat composition. Gynecol. Endocrinol. 23, 99–104 (2007)PubMedCrossRef
Metadaten
Titel
The effect of chronic immobilization stress on leptin signaling in the ovariectomized (OVX) rat
verfasst von
Darwin O. Larco
Danette F. Cruthirds
Michael J. Weiser
Robert J. Handa
T. John Wu
Publikationsdatum
01.12.2012
Verlag
Springer US
Erschienen in
Endocrine / Ausgabe 3/2012
Print ISSN: 1355-008X
Elektronische ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-012-9716-x

Weitere Artikel der Ausgabe 3/2012

Endocrine 3/2012 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.