Skip to main content
Top
Published in: Endocrine 1/2018

01-10-2018 | Original Article

Downregulation of leptin receptor and kisspeptin/GPR54 in the murine hypothalamus contributes to male hypogonadism caused by high-fat diet-induced obesity

Authors: Lingling Zhai, Jian Zhao, Yiming Zhu, Qiannan Liu, Wenhua Niu, Chengyin Liu, Yi Wang

Published in: Endocrine | Issue 1/2018

Login to get access

Abstract

Purpose

Obesity may lead to male hypogonadism, the underlying mechanism of which remains unclear. In the present study, we established a murine model of male hypogonadism caused by high-fat diet-induced obesity to verify the following hypotheses: 1) an increased leptin level may be related to decreased secretion of GnRH in obese males, and 2) repression of kisspeptin/GPR54 in the hypothalamus, which is associated with increased leptin levels, may account for the decreased secretion of GnRH and be involved in secondary hypogonadism (SH) in obese males.

Methods

Male mice were fed high-fat diet for 19 weeks and divided by body weight gain into diet-induced obesity (DIO) and diet-induced obesity resistant (DIO-R) group. The effect of obesity on the reproductive organs in male mice was observed by measuring sperm count and spermatozoid motility, relative to testis and epididymis weight, testosterone levels, and pathologic changes. Leptin, testosterone, estrogen, and LH in serum were detected by ELISA method. Leptin receptor (Ob-R), Kiss1, GPR54, and GnRH mRNA were measured by real-time PCR in the hypothalamus. Expression of kisspeptin and Ob-R protein was determined by Western blotting. Expression of GnRH and GPR54 protein was determined by immunohistochemical analysis.

Results

We found that diet-induced obesity decreased spermatozoid motility, testis and epididymis relative coefficients, and plasma testosterone and luteinizing hormone levels. An increased number and volume of lipid droplets in Leydig cells were observed in the DIO group compared to the control group. Significantly, higher serum leptin levels were found in the DIO and DIO-R groups. The DIO and DIO-R groups showed significant downregulation of the GnRH, Kiss1, GPR54, and Ob-R genes. We also found decreased levels of GnRH, kisspeptin, GPR54, and Ob-R protein in the DIO and DIO-R groups.

Conclusions

These lines of evidence suggest that downregulation of Ob-R and kisspeptin/GPR54 in the murine hypothalamus may contribute to male hypogonadism caused by high-fat diet-induced obesity.
Appendix
Available only for authorised users
Literature
1.
go back to reference M. Ng, T. Fleming, M. Robinson, B. Thomson, N. Graetz, C. Margono et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766–81 (2014)CrossRefPubMedPubMedCentral M. Ng, T. Fleming, M. Robinson, B. Thomson, N. Graetz, C. Margono et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766–81 (2014)CrossRefPubMedPubMedCentral
2.
go back to reference J.Y. Hwang, H.J. Lee, M.J. Go, H.B. Jang, S.I. Park, B.J. Kim, H.J. Lee, An integrative study identifies KCNC2 as a novel predisposing factor for childhood obesity and the risk of diabetes in the Korean population. Sci. Rep. 6, 33043 (2016)CrossRefPubMedPubMedCentral J.Y. Hwang, H.J. Lee, M.J. Go, H.B. Jang, S.I. Park, B.J. Kim, H.J. Lee, An integrative study identifies KCNC2 as a novel predisposing factor for childhood obesity and the risk of diabetes in the Korean population. Sci. Rep. 6, 33043 (2016)CrossRefPubMedPubMedCentral
3.
go back to reference M. Horikoshi, R.N. Beaumont, F.R. Day, N.M. Warrington, M.N. Kooijman, J. Fernandez-Tajes et al. Genome-wide associations for birth weight and correlations with adult disease. Nature 538, 248–52 (2016)CrossRefPubMedPubMedCentral M. Horikoshi, R.N. Beaumont, F.R. Day, N.M. Warrington, M.N. Kooijman, J. Fernandez-Tajes et al. Genome-wide associations for birth weight and correlations with adult disease. Nature 538, 248–52 (2016)CrossRefPubMedPubMedCentral
4.
go back to reference D.M. Allcock, M.J. Gardner, J.R. Sowers, Relation between childhood obesity and adult cardiovascular risk. Int J. Pediatr. Endocrinol. 2009, 10887 (2009)CrossRef D.M. Allcock, M.J. Gardner, J.R. Sowers, Relation between childhood obesity and adult cardiovascular risk. Int J. Pediatr. Endocrinol. 2009, 10887 (2009)CrossRef
5.
go back to reference L.J. Lloyd, S.C. Langley-Evans, S. McMullen, Childhood obesity and risk of the adult metabolic syndrome: a systematic review. Int J. Obes. (Lond.) 36, 1–11 (2012)CrossRef L.J. Lloyd, S.C. Langley-Evans, S. McMullen, Childhood obesity and risk of the adult metabolic syndrome: a systematic review. Int J. Obes. (Lond.) 36, 1–11 (2012)CrossRef
6.
go back to reference J.L. Baker, L.W. Olsen, T.I. Sørensen, Childhood body-mass index and the risk of coronary heart disease in adulthood. N. Engl. J. Med 357, 2329–37 (2007)CrossRefPubMedPubMedCentral J.L. Baker, L.W. Olsen, T.I. Sørensen, Childhood body-mass index and the risk of coronary heart disease in adulthood. N. Engl. J. Med 357, 2329–37 (2007)CrossRefPubMedPubMedCentral
8.
go back to reference Y. Wang, Is obesity associated with early sexual maturation? A comparison of the association in American boys versus girls. Pediatrics 110, 903–10 (2002)CrossRefPubMed Y. Wang, Is obesity associated with early sexual maturation? A comparison of the association in American boys versus girls. Pediatrics 110, 903–10 (2002)CrossRefPubMed
9.
go back to reference J.E. Blundell, J.L. Baker, E. Boyland, E. Blaak, J. Charzewska, S. de Henauw, G. Frühbeck, M. Gonzalez-Gross, J. Hebebrand, L. Holm, V. Kriaucioniene, L. Lissner, J.M. Oppert, K. Schindler, A.M. Silva, E. Woodward, Variations in the prevalence of obesity among European countries and a consideration of possible causes. Obes. Facts 10, 25–37 (2017)CrossRefPubMedPubMedCentral J.E. Blundell, J.L. Baker, E. Boyland, E. Blaak, J. Charzewska, S. de Henauw, G. Frühbeck, M. Gonzalez-Gross, J. Hebebrand, L. Holm, V. Kriaucioniene, L. Lissner, J.M. Oppert, K. Schindler, A.M. Silva, E. Woodward, Variations in the prevalence of obesity among European countries and a consideration of possible causes. Obes. Facts 10, 25–37 (2017)CrossRefPubMedPubMedCentral
10.
go back to reference C.L. Ogden, M.D. Carroll, C.D. Fryar, K.M. Flegal, Prevalence of obesity among adults and youth: United States, 2011–2014. (US Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Health Statistics, 2015). C.L. Ogden, M.D. Carroll, C.D. Fryar, K.M. Flegal, Prevalence of obesity among adults and youth: United States, 2011–2014. (US Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Health Statistics, 2015).
11.
go back to reference National Health and Family Planning Commission of the PRC, 2014 Report on Chinese resident’s chronic disease and nutrition. (2015). National Health and Family Planning Commission of the PRC, 2014 Report on Chinese resident’s chronic disease and nutrition. (2015).
12.
go back to reference W. Kiess, I.V. Wagner, J. Kratzsch, A. Körner, Male obesity. Endocrinol. Metab. Clin. North Am. 44, 761–72 (2015)CrossRefPubMed W. Kiess, I.V. Wagner, J. Kratzsch, A. Körner, Male obesity. Endocrinol. Metab. Clin. North Am. 44, 761–72 (2015)CrossRefPubMed
13.
go back to reference S.S. Du Plessis, S. Cabler, D.A. McAlister, E. Sabanegh, A. Agarwal, The effect of obesity on sperm disorders and male infertility. Nat. Rev. Urol. 7, 153–61 (2010)CrossRefPubMed S.S. Du Plessis, S. Cabler, D.A. McAlister, E. Sabanegh, A. Agarwal, The effect of obesity on sperm disorders and male infertility. Nat. Rev. Urol. 7, 153–61 (2010)CrossRefPubMed
14.
go back to reference N.O. Palmer, H.W. Bakos, T. Fullston, M. Lane, Impact of obesity on male fertility, sperm function and molecular composition. Spermatogenesis 2, 253–63 (2012)CrossRefPubMedPubMedCentral N.O. Palmer, H.W. Bakos, T. Fullston, M. Lane, Impact of obesity on male fertility, sperm function and molecular composition. Spermatogenesis 2, 253–63 (2012)CrossRefPubMedPubMedCentral
15.
go back to reference G. Ciocca, E. Limoncin, E. Carosa, S. Di Sante, G.L. Gravina, D. Mollaioli, D. Gianfrilli, A. Lenzi, E.A. Jannini, Is Testosterone a Food for the Brain? Sex. Med. Rev. 4, 15–25 (2016)CrossRefPubMed G. Ciocca, E. Limoncin, E. Carosa, S. Di Sante, G.L. Gravina, D. Mollaioli, D. Gianfrilli, A. Lenzi, E.A. Jannini, Is Testosterone a Food for the Brain? Sex. Med. Rev. 4, 15–25 (2016)CrossRefPubMed
16.
go back to reference J.T. George, R.P. Millar, R.A. Anderson, Hypothesis: kisspeptin mediates male hypogonadism in obesity and type 2 diabetes. Neuroendocrinology 91, 302–7 (2010)CrossRefPubMed J.T. George, R.P. Millar, R.A. Anderson, Hypothesis: kisspeptin mediates male hypogonadism in obesity and type 2 diabetes. Neuroendocrinology 91, 302–7 (2010)CrossRefPubMed
17.
go back to reference A. Olivares, J.P. Méndez, E. Zambrano, M. Cárdenas, A. Tovar, G. Perera-Marín, A. Ulloa-Aguirre, Reproductive axis function and gonadotropin microheterogeneity in a male rat model of diet-induced obesity. Gen. Comp. Endocrinol. 166, 356–64 (2010)CrossRefPubMed A. Olivares, J.P. Méndez, E. Zambrano, M. Cárdenas, A. Tovar, G. Perera-Marín, A. Ulloa-Aguirre, Reproductive axis function and gonadotropin microheterogeneity in a male rat model of diet-induced obesity. Gen. Comp. Endocrinol. 166, 356–64 (2010)CrossRefPubMed
18.
go back to reference S.R. Ojeda, C. Dubay, A. Lomniczi, G. Kaidar, V. Matagne, U.S. Sandau, G.A. Dissen, Gene networks and the neuroendocrine regulation of puberty. Mol. Cell Endocrinol. 324, 3–11 (2010)CrossRefPubMed S.R. Ojeda, C. Dubay, A. Lomniczi, G. Kaidar, V. Matagne, U.S. Sandau, G.A. Dissen, Gene networks and the neuroendocrine regulation of puberty. Mol. Cell Endocrinol. 324, 3–11 (2010)CrossRefPubMed
19.
go back to reference M.A. Sánchez-Garrido, F. Ruiz-Pino, M. Manfredi-Lozano, S. Leon, D. Garcia-Galiano, J.P. Castaño, R.M. Luque, A. Romero-Ruiz, J.M. Castellano, C. Diéguez, L. Pinilla, M. Tena-Sempere, Obesity-induced hypogonadism in the male: premature reproductive neuroendocrine senescence and contribution of Kiss1-mediated mechanisms. Endocrinology 155, 1067–79 (2014)CrossRefPubMed M.A. Sánchez-Garrido, F. Ruiz-Pino, M. Manfredi-Lozano, S. Leon, D. Garcia-Galiano, J.P. Castaño, R.M. Luque, A. Romero-Ruiz, J.M. Castellano, C. Diéguez, L. Pinilla, M. Tena-Sempere, Obesity-induced hypogonadism in the male: premature reproductive neuroendocrine senescence and contribution of Kiss1-mediated mechanisms. Endocrinology 155, 1067–79 (2014)CrossRefPubMed
20.
go back to reference Y.J. Rhie, Kisspeptin/G protein-coupled receptor-54 system as an essential gatekeeper of pubertal development. Ann. Pediatr. Endocrinol. Metab. 18, 55–59 (2013)CrossRefPubMedPubMedCentral Y.J. Rhie, Kisspeptin/G protein-coupled receptor-54 system as an essential gatekeeper of pubertal development. Ann. Pediatr. Endocrinol. Metab. 18, 55–59 (2013)CrossRefPubMedPubMedCentral
21.
go back to reference D. Garcia-Galiano, S.J. Allen, C.F. Elias, Role of the adipocyte-derived hormone leptin in reproductive control. Horm. Mol. Biol. Clin. Investig. 19, 141–9 (2014)PubMedPubMedCentral D. Garcia-Galiano, S.J. Allen, C.F. Elias, Role of the adipocyte-derived hormone leptin in reproductive control. Horm. Mol. Biol. Clin. Investig. 19, 141–9 (2014)PubMedPubMedCentral
22.
go back to reference J. Wauman, J. Tavernier, Leptin receptor signaling: pathways to leptin resistance. Front. Biosci. 16, 2771–93 (2011)CrossRef J. Wauman, J. Tavernier, Leptin receptor signaling: pathways to leptin resistance. Front. Biosci. 16, 2771–93 (2011)CrossRef
23.
go back to reference M. Mahmoodzadeh Sagheb, N. Azarpira, R. Yaghobi, The effect of leptin and adiponectin on KiSS-1 and KissR mRNA expression in rat islets of langerhans and CRI-D2 cell line. Int J. Endocrinol. Metab. 12, e15297 (2014)CrossRefPubMedPubMedCentral M. Mahmoodzadeh Sagheb, N. Azarpira, R. Yaghobi, The effect of leptin and adiponectin on KiSS-1 and KissR mRNA expression in rat islets of langerhans and CRI-D2 cell line. Int J. Endocrinol. Metab. 12, e15297 (2014)CrossRefPubMedPubMedCentral
24.
go back to reference L. Pinilla, E. Aguilar, C. Dieguez, R.P. Millar, M. Tena-Sempere, Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol. Rev. 92, 1235–316 (2012)CrossRefPubMed L. Pinilla, E. Aguilar, C. Dieguez, R.P. Millar, M. Tena-Sempere, Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol. Rev. 92, 1235–316 (2012)CrossRefPubMed
26.
go back to reference Y. Iwasaki, Y. Maejima, S. Suyama, M. Yoshida, T. Arai, K. Katsurada, P. Kumari, H. Nakabayashi, M. Kakei, T. Yada, Peripheral oxytocin activates vagal afferent neurons to suppress feeding in normal and leptin-resistant mice: a route for ameliorating hyperphagia and obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308, R360–R369 (2015)CrossRefPubMed Y. Iwasaki, Y. Maejima, S. Suyama, M. Yoshida, T. Arai, K. Katsurada, P. Kumari, H. Nakabayashi, M. Kakei, T. Yada, Peripheral oxytocin activates vagal afferent neurons to suppress feeding in normal and leptin-resistant mice: a route for ameliorating hyperphagia and obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 308, R360–R369 (2015)CrossRefPubMed
27.
go back to reference T. Kusakabe, K. Ebihara, T. Sakai, L. Miyamoto, D. Aotani, Y. Yamamoto, S. Yamamoto-Kataoka, M. Aizawa-Abe, J. Fujikura, K. Hosoda, K. Nakao, Amylin improves the effect of leptin on insulin sensitivity in leptin-resistant diet-induced obese mice. Am. J. Physiol. Endocrinol. Metab. 302, E924–E931 (2012)CrossRefPubMed T. Kusakabe, K. Ebihara, T. Sakai, L. Miyamoto, D. Aotani, Y. Yamamoto, S. Yamamoto-Kataoka, M. Aizawa-Abe, J. Fujikura, K. Hosoda, K. Nakao, Amylin improves the effect of leptin on insulin sensitivity in leptin-resistant diet-induced obese mice. Am. J. Physiol. Endocrinol. Metab. 302, E924–E931 (2012)CrossRefPubMed
28.
go back to reference J. Zhao, L. Zhai, Z. Liu, S. Wu, L. Xu, Leptin level and oxidative stress contribute to obesity-induced low testosterone in murine testicular tissue. Oxid. Med Cell Longev. 2014, 190945 (2014)PubMedPubMedCentral J. Zhao, L. Zhai, Z. Liu, S. Wu, L. Xu, Leptin level and oxidative stress contribute to obesity-induced low testosterone in murine testicular tissue. Oxid. Med Cell Longev. 2014, 190945 (2014)PubMedPubMedCentral
29.
go back to reference B.E. Levin, R.E. Keesey, Defense of differing body weight set points in diet-induced obese and resistant rats. Am. J. Physiol. 274, R412–R419 (1998)PubMed B.E. Levin, R.E. Keesey, Defense of differing body weight set points in diet-induced obese and resistant rats. Am. J. Physiol. 274, R412–R419 (1998)PubMed
30.
go back to reference WHO. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction. (Cambridge University Press, Cambridge, 1999) WHO. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction. (Cambridge University Press, Cambridge, 1999)
31.
go back to reference K.J. Teerds, D.G. de Rooij, J. Keijer, Functional relationship between obesity and male reproduction: from humans to animal models. Hum. Reprod. Update 17, 667–83 (2011)CrossRefPubMed K.J. Teerds, D.G. de Rooij, J. Keijer, Functional relationship between obesity and male reproduction: from humans to animal models. Hum. Reprod. Update 17, 667–83 (2011)CrossRefPubMed
32.
33.
go back to reference G.Y. Bédécarrats, Control of the reproductive axis: balancing act between stimulatory and inhibitory inputs. Poult. Sci. 94, 810–5 (2015)CrossRefPubMed G.Y. Bédécarrats, Control of the reproductive axis: balancing act between stimulatory and inhibitory inputs. Poult. Sci. 94, 810–5 (2015)CrossRefPubMed
34.
go back to reference P. Dandona, S. Dhindsa, A. Chaudhuri, V. Bhatia, S. Topiwala, P. Mohanty, Hypogonadotrophic hypogonadism in type 2 diabetes, obesity and the metabolic syndrome. Curr. Mol. Med 8, 816–28 (2008)CrossRefPubMed P. Dandona, S. Dhindsa, A. Chaudhuri, V. Bhatia, S. Topiwala, P. Mohanty, Hypogonadotrophic hypogonadism in type 2 diabetes, obesity and the metabolic syndrome. Curr. Mol. Med 8, 816–28 (2008)CrossRefPubMed
35.
go back to reference Y. Zhang, R. Proenca, M. Maffei, M. Barone, L. Leopold, J.M. Friedman, Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–32 (1994)CrossRefPubMed Y. Zhang, R. Proenca, M. Maffei, M. Barone, L. Leopold, J.M. Friedman, Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–32 (1994)CrossRefPubMed
36.
go back to reference C.R. McCartney, K.A. Prendergast, S.K. Blank, K.D. Helm, S. Chhabra, J.C. Marshall, Maturation of luteinizing hormone (gonadotropin-releasing hormone) secretion across puberty: evidence for altered regulation in obese peripubertal girls. J. Clin. Endocrinol. Metab. 94, 56–66 (2009)CrossRefPubMed C.R. McCartney, K.A. Prendergast, S.K. Blank, K.D. Helm, S. Chhabra, J.C. Marshall, Maturation of luteinizing hormone (gonadotropin-releasing hormone) secretion across puberty: evidence for altered regulation in obese peripubertal girls. J. Clin. Endocrinol. Metab. 94, 56–66 (2009)CrossRefPubMed
37.
go back to reference F.F. Casanueva, C. Dieguez, Neuroendocrine regulation and actions of leptin. Front Neuroendocrinol. 20, 317–63 (1999)CrossRefPubMed F.F. Casanueva, C. Dieguez, Neuroendocrine regulation and actions of leptin. Front Neuroendocrinol. 20, 317–63 (1999)CrossRefPubMed
38.
go back to reference M.J. Cunningham, D.K. Clifton, R.A. Steiner, Leptin’s actions on the reproductive axis: perspectives and mechanisms. Biol. Reprod. 60, 216–22 (1999)CrossRefPubMed M.J. Cunningham, D.K. Clifton, R.A. Steiner, Leptin’s actions on the reproductive axis: perspectives and mechanisms. Biol. Reprod. 60, 216–22 (1999)CrossRefPubMed
39.
go back to reference J. Roa, E. Aguilar, C. Dieguez, L. Pinilla, M. Tena-Sempere, New frontiers in kisspeptin/GPR54 physiology as fundamental gatekeepers of reproductive function. Front Neuroendocrinol. 29, 48–69 (2008)CrossRefPubMed J. Roa, E. Aguilar, C. Dieguez, L. Pinilla, M. Tena-Sempere, New frontiers in kisspeptin/GPR54 physiology as fundamental gatekeepers of reproductive function. Front Neuroendocrinol. 29, 48–69 (2008)CrossRefPubMed
40.
go back to reference J.M. Castellano, V.M. Navarro, R. Fernández-Fernández, J. Roa, E. Vigo, R. Pineda, C. Dieguez, E. Aguilar, L. Pinilla, M. Tena-Sempere, Expression of hypothalamic KiSS-1 system and rescue of defective gonadotropic responses by kisspeptin in streptozotocin-induced diabetic male rats. Diabetes 55, 2602–10 (2006)CrossRefPubMed J.M. Castellano, V.M. Navarro, R. Fernández-Fernández, J. Roa, E. Vigo, R. Pineda, C. Dieguez, E. Aguilar, L. Pinilla, M. Tena-Sempere, Expression of hypothalamic KiSS-1 system and rescue of defective gonadotropic responses by kisspeptin in streptozotocin-induced diabetic male rats. Diabetes 55, 2602–10 (2006)CrossRefPubMed
42.
go back to reference A.K. Roseweir, A.S. Kauffman, J.T. Smith, K.A. Guerriero, K. Morgan, J. Pielecka-Fortuna, R. Pineda, M.L. Gottsch, M. Tena-Sempere, S.M. Moenter, E. Terasawa, I.J. Clarke, R.A. Steiner, R.P. Millar, Discovery of potent kisspeptin antagonists delineate physiological mechanisms of gonadotropin regulation. J. Neurosci. 29, 3920–9 (2009)CrossRefPubMedPubMedCentral A.K. Roseweir, A.S. Kauffman, J.T. Smith, K.A. Guerriero, K. Morgan, J. Pielecka-Fortuna, R. Pineda, M.L. Gottsch, M. Tena-Sempere, S.M. Moenter, E. Terasawa, I.J. Clarke, R.A. Steiner, R.P. Millar, Discovery of potent kisspeptin antagonists delineate physiological mechanisms of gonadotropin regulation. J. Neurosci. 29, 3920–9 (2009)CrossRefPubMedPubMedCentral
43.
go back to reference J.T. George, R.A. Anderson, R.P. Millar, Kisspeptin-10 stimulation of gonadotrophin secretion in women is modulated by sex steroid feedback. Hum. Reprod. 27, 3552–9 (2012)CrossRefPubMed J.T. George, R.A. Anderson, R.P. Millar, Kisspeptin-10 stimulation of gonadotrophin secretion in women is modulated by sex steroid feedback. Hum. Reprod. 27, 3552–9 (2012)CrossRefPubMed
44.
go back to reference J.M. Castellano, J. Roa, R.M. Luque, C. Dieguez, E. Aguilar, L. Pinilla, M. Tena-Sempere, KiSS-1/kisspeptins and the metabolic control of reproduction: physiologic roles and putative physiopathological implications. Peptides 30, 139–45 (2009)CrossRefPubMed J.M. Castellano, J. Roa, R.M. Luque, C. Dieguez, E. Aguilar, L. Pinilla, M. Tena-Sempere, KiSS-1/kisspeptins and the metabolic control of reproduction: physiologic roles and putative physiopathological implications. Peptides 30, 139–45 (2009)CrossRefPubMed
45.
go back to reference R.M. Luque, R.D. Kinema, M. Tena-Sempere, Regulation of hypothalamic expression of KiSS-1 and GPR54 genes by metabolic factors: analyses using mouse models and a cell line. Endocrinology 148, 4601–11 (2007)CrossRefPubMed R.M. Luque, R.D. Kinema, M. Tena-Sempere, Regulation of hypothalamic expression of KiSS-1 and GPR54 genes by metabolic factors: analyses using mouse models and a cell line. Endocrinology 148, 4601–11 (2007)CrossRefPubMed
46.
go back to reference J.T. Smith, B.V. Acohido, D.K. Clifton, R.A. Steiner, KiSS-1 neurones are direct targets for leptin in the ob/ob mouse. J. Neuroendocrinol. 18, 298–303 (2006)CrossRefPubMed J.T. Smith, B.V. Acohido, D.K. Clifton, R.A. Steiner, KiSS-1 neurones are direct targets for leptin in the ob/ob mouse. J. Neuroendocrinol. 18, 298–303 (2006)CrossRefPubMed
47.
go back to reference J. Wilsey, P.J. Scarpace, Caloric restriction reverses the deficits in leptin receptor protein and leptin signaling capacity associated with diet-induced obesity: role of leptin in the regulation of hypothalamic long-form leptin receptor expression. J. Endocrinol. 181, 297–306 (2004)CrossRefPubMed J. Wilsey, P.J. Scarpace, Caloric restriction reverses the deficits in leptin receptor protein and leptin signaling capacity associated with diet-induced obesity: role of leptin in the regulation of hypothalamic long-form leptin receptor expression. J. Endocrinol. 181, 297–306 (2004)CrossRefPubMed
48.
go back to reference Y.M. Han, G.M. Kang, K. Byun, H.W. Ko, J. Kim, M.S. Shin, H.K. Kim, S.Y. Gil, J.H. Yu, B. Lee, M.S. Kim, Leptin-promoted cilia assembly is critical for normal energy balance. J. Clin. Invest 124, 2193–7 (2014)CrossRefPubMedPubMedCentral Y.M. Han, G.M. Kang, K. Byun, H.W. Ko, J. Kim, M.S. Shin, H.K. Kim, S.Y. Gil, J.H. Yu, B. Lee, M.S. Kim, Leptin-promoted cilia assembly is critical for normal energy balance. J. Clin. Invest 124, 2193–7 (2014)CrossRefPubMedPubMedCentral
49.
go back to reference M. Schneeberger, M.O. Dietrich, D. Sebastián, M. Imbernón, C. Castaño, A. Garcia, Y. Esteban, A. Gonzalez-Franquesa, I.C. Rodríguez, A. Bortolozzi, P.M. Garcia-Roves, R. Gomis, R. Nogueiras, T.L. Horvath, A. Zorzano, M. Claret, Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155, 172–87 (2013)CrossRefPubMed M. Schneeberger, M.O. Dietrich, D. Sebastián, M. Imbernón, C. Castaño, A. Garcia, Y. Esteban, A. Gonzalez-Franquesa, I.C. Rodríguez, A. Bortolozzi, P.M. Garcia-Roves, R. Gomis, R. Nogueiras, T.L. Horvath, A. Zorzano, M. Claret, Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155, 172–87 (2013)CrossRefPubMed
50.
go back to reference M.G. Jr Myers, S.B. Heymsfield, C. Haft, B.B. Kahn, M. Laughlin, R.L. Leibel, M.H. Tschöp, J.A. Yanovski, Challenges and opportunities of defining clinical leptin resistance. Cell Metab. 15, 150–6 (2012)CrossRefPubMedPubMedCentral M.G. Jr Myers, S.B. Heymsfield, C. Haft, B.B. Kahn, M. Laughlin, R.L. Leibel, M.H. Tschöp, J.A. Yanovski, Challenges and opportunities of defining clinical leptin resistance. Cell Metab. 15, 150–6 (2012)CrossRefPubMedPubMedCentral
51.
go back to reference J. Clarkson, A.E. Herbison, Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons. Endocrinology 12, 5817–25 (2006)CrossRef J. Clarkson, A.E. Herbison, Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons. Endocrinology 12, 5817–25 (2006)CrossRef
52.
go back to reference J.T. Smith, H.M. Dungan, E.A. Stoll, M.L. Gottsch, R.E. Braun, S.M. Eacker, D.K. Clifton, R.A. Steiner, Differential regulation of KiSS-1 mRNA expression by sex steroids in the brain of the male mouse. Endocrinology 7, 2976–84 (2005)CrossRef J.T. Smith, H.M. Dungan, E.A. Stoll, M.L. Gottsch, R.E. Braun, S.M. Eacker, D.K. Clifton, R.A. Steiner, Differential regulation of KiSS-1 mRNA expression by sex steroids in the brain of the male mouse. Endocrinology 7, 2976–84 (2005)CrossRef
53.
go back to reference K. Backholer, J. Smith, I.J. Clarke, Melanocortins may stimulate reproduction by activating orexin neurons in the dorsomedial hypothalamus and kisspeptin neurons in the preoptic area of the ewe. Endocrinology 12, 5488–97 (2009)CrossRef K. Backholer, J. Smith, I.J. Clarke, Melanocortins may stimulate reproduction by activating orexin neurons in the dorsomedial hypothalamus and kisspeptin neurons in the preoptic area of the ewe. Endocrinology 12, 5488–97 (2009)CrossRef
54.
go back to reference L. Pinilla, E. Aguilar, C. Dieguez, R.P. Millar, M. Tena-Sempere, Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol. Rev. 3, 1235–316 (2012)CrossRef L. Pinilla, E. Aguilar, C. Dieguez, R.P. Millar, M. Tena-Sempere, Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol. Rev. 3, 1235–316 (2012)CrossRef
55.
go back to reference Z. Javed, U. Qamar, T. Sathyapalan, The role of kisspeptin signalling in the hypothalamic-pituitary-gonadal axis-current perspective. Endokrynol. Pol. 6, 534–47 (2015)CrossRef Z. Javed, U. Qamar, T. Sathyapalan, The role of kisspeptin signalling in the hypothalamic-pituitary-gonadal axis-current perspective. Endokrynol. Pol. 6, 534–47 (2015)CrossRef
56.
go back to reference R. Chianese, V. Ciaramella, S. Fasano, R. Pierantoni, R. Meccariello, Kisspeptin drives germ cell progression in the anuran amphibian Pelophylax esculentus: a study carried out in ex vivo testes. Gen. Comp. Endocrinol. 211, 81–91 (2015)CrossRefPubMed R. Chianese, V. Ciaramella, S. Fasano, R. Pierantoni, R. Meccariello, Kisspeptin drives germ cell progression in the anuran amphibian Pelophylax esculentus: a study carried out in ex vivo testes. Gen. Comp. Endocrinol. 211, 81–91 (2015)CrossRefPubMed
57.
58.
go back to reference R. Meccariello, R. Chianese, T. Chioccarelli, V. Ciaramella, S. Fasano, R. Pierantoni, G. Cobellis, Intra-testicular signals regulate germ cell progression and production of qualitatively mature spermatozoa in vertebrates. Front. Endocrinol. 5, 69 (2014) R. Meccariello, R. Chianese, T. Chioccarelli, V. Ciaramella, S. Fasano, R. Pierantoni, G. Cobellis, Intra-testicular signals regulate germ cell progression and production of qualitatively mature spermatozoa in vertebrates. Front. Endocrinol. 5, 69 (2014)
59.
60.
go back to reference A.M. Isidori, M. Caprio, F. Strollo, C. Moretti, G. Frajese, A. Isidori, A. Fabbri, Leptin and androgens in male obesity: Evidence for leptin contribution to reduced androgen levels. J. Clin. Endocrinol. Metab. 84, 3673–80 (1999)PubMed A.M. Isidori, M. Caprio, F. Strollo, C. Moretti, G. Frajese, A. Isidori, A. Fabbri, Leptin and androgens in male obesity: Evidence for leptin contribution to reduced androgen levels. J. Clin. Endocrinol. Metab. 84, 3673–80 (1999)PubMed
61.
go back to reference H. Zhang, K. Taya, K. Nagaoka, M. Yoshida, G. Watanabe, Neonatal exposure to 17α-ethynyl estradiol (EE) disrupts follicle development and reproductive hormone profiles in female rats. Toxicol. Lett. 276, 92–99 (2017)CrossRefPubMed H. Zhang, K. Taya, K. Nagaoka, M. Yoshida, G. Watanabe, Neonatal exposure to 17α-ethynyl estradiol (EE) disrupts follicle development and reproductive hormone profiles in female rats. Toxicol. Lett. 276, 92–99 (2017)CrossRefPubMed
62.
go back to reference E. Pruszyńska-Oszmałek, P.A. Kołodziejski, M. Sassek, J.H. Sliwowska, Kisspeptin-10 inhibits proliferation and regulates lipolysis and lipogenesis processes in 3T3-L1 cells and isolated rat adipocytes. Endocrine 56, 54–64 (2017)CrossRefPubMed E. Pruszyńska-Oszmałek, P.A. Kołodziejski, M. Sassek, J.H. Sliwowska, Kisspeptin-10 inhibits proliferation and regulates lipolysis and lipogenesis processes in 3T3-L1 cells and isolated rat adipocytes. Endocrine 56, 54–64 (2017)CrossRefPubMed
63.
go back to reference M. Dudek, P.A. Kołodziejski, E. Pruszyńska-Oszmałek, M. Sassek, K. Ziarniak, K.W. Nowak, J.H. Sliwowska, Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the hypothalamic-pituitary-gonadal (HPG) axis and peripheral organs (fat, pancreas and liver) in male rats. Neuropeptides 56, 41–9 (2016)CrossRefPubMed M. Dudek, P.A. Kołodziejski, E. Pruszyńska-Oszmałek, M. Sassek, K. Ziarniak, K.W. Nowak, J.H. Sliwowska, Effects of high-fat diet-induced obesity and diabetes on Kiss1 and GPR54 expression in the hypothalamic-pituitary-gonadal (HPG) axis and peripheral organs (fat, pancreas and liver) in male rats. Neuropeptides 56, 41–9 (2016)CrossRefPubMed
65.
go back to reference W.J. Song, P. Mondal, A. Wolfe, L.C. Alonso, R. Stamateris, B.W. Ong, O.C. Lim, K.S. Yang, S. Radovick, H.J. Novaira, E.A. Farber, C.R. Farber, S.D. Turner, M.A. Hussain, Glucagon regulates hepatic kisspeptin to impair insulin secretion. Cell Metab. 19, 667–81 (2014)CrossRefPubMedPubMedCentral W.J. Song, P. Mondal, A. Wolfe, L.C. Alonso, R. Stamateris, B.W. Ong, O.C. Lim, K.S. Yang, S. Radovick, H.J. Novaira, E.A. Farber, C.R. Farber, S.D. Turner, M.A. Hussain, Glucagon regulates hepatic kisspeptin to impair insulin secretion. Cell Metab. 19, 667–81 (2014)CrossRefPubMedPubMedCentral
Metadata
Title
Downregulation of leptin receptor and kisspeptin/GPR54 in the murine hypothalamus contributes to male hypogonadism caused by high-fat diet-induced obesity
Authors
Lingling Zhai
Jian Zhao
Yiming Zhu
Qiannan Liu
Wenhua Niu
Chengyin Liu
Yi Wang
Publication date
01-10-2018
Publisher
Springer US
Published in
Endocrine / Issue 1/2018
Print ISSN: 1355-008X
Electronic ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-018-1646-9

Other articles of this Issue 1/2018

Endocrine 1/2018 Go to the issue