Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2019

Open Access 01-12-2019 | Review

Reproductive functions of Kisspeptin/KISS1R Systems in the Periphery

Authors: Yubin Cao, Zeping Li, Wenyu Jiang, Yan Ling, Haibin Kuang

Published in: Reproductive Biology and Endocrinology | Issue 1/2019

Login to get access

Abstract

Kisspeptin and its G protein-coupled receptor KISS1R play key roles in mammalian reproduction due to their involvement in the onset of puberty and control of the hypothalamic-pituitary-gonadal axis. However, recent studies have indicated a potential role of extra-hypothalamic kisspeptin in reproductive function. Here, we summarize recent advances in our understanding of the physiological significance of kisspeptin/KISS1R in the peripheral reproductive system (including the ovary, testis, uterus, and placenta) and the potential role of kisspeptin/KISS1R in reproductive diseases. A comprehensive understanding of the expression, function, and potential molecular mechanisms of kisspeptin/KISS1R in the peripheral reproductive system will contribute to the diagnosis, treatment and prevention of reproductive diseases.
Literature
1.
go back to reference Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev. 2012;92(3):1235–316.PubMedCrossRef Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev. 2012;92(3):1235–316.PubMedCrossRef
2.
go back to reference Herbison AE. Control of puberty onset and fertility by gonadotropin-releasing hormone neurons. Nat Rev Endocrinol. 2016;12(8):452–66.PubMedCrossRef Herbison AE. Control of puberty onset and fertility by gonadotropin-releasing hormone neurons. Nat Rev Endocrinol. 2016;12(8):452–66.PubMedCrossRef
3.
go back to reference Shahab M, Mastronardi C, Seminara SB, Crowley WF, Ojeda SR, Plant TM. Increased hypothalamic GPR54 signaling: a potential mechanism for initiation of puberty in primates. Proc Natl Acad Sci U S A. 2005;102(6):2129–34.PubMedPubMedCentralCrossRef Shahab M, Mastronardi C, Seminara SB, Crowley WF, Ojeda SR, Plant TM. Increased hypothalamic GPR54 signaling: a potential mechanism for initiation of puberty in primates. Proc Natl Acad Sci U S A. 2005;102(6):2129–34.PubMedPubMedCentralCrossRef
4.
go back to reference Terao Y, Kumano S, Takatsu Y, Hattori M, Nishimura A, Ohtaki T, et al. Expression of KiSS-1, a metastasis suppressor gene, in trophoblast giant cells of the rat placenta. Biochim Biophys Acta. 2004;1678(2–3):102–10.PubMedCrossRef Terao Y, Kumano S, Takatsu Y, Hattori M, Nishimura A, Ohtaki T, et al. Expression of KiSS-1, a metastasis suppressor gene, in trophoblast giant cells of the rat placenta. Biochim Biophys Acta. 2004;1678(2–3):102–10.PubMedCrossRef
6.
go back to reference Cejudo Roman A, Pinto FM, Dorta I, Almeida TA, Hernandez M, Illanes M, et al. Analysis of the expression of neurokinin B, kisspeptin, and their cognate receptors NK3R and KISS1R in the human female genital tract. Fertil Steril. 2012;97(5):1213–9.PubMedCrossRef Cejudo Roman A, Pinto FM, Dorta I, Almeida TA, Hernandez M, Illanes M, et al. Analysis of the expression of neurokinin B, kisspeptin, and their cognate receptors NK3R and KISS1R in the human female genital tract. Fertil Steril. 2012;97(5):1213–9.PubMedCrossRef
7.
go back to reference Hu KL, Zhao H, Chang HM, Yu Y, Qiao J. Kisspeptin/Kisspeptin receptor system in the ovary. Front Endocrinol (Lausanne). 2017;8:365.PubMedCrossRef Hu KL, Zhao H, Chang HM, Yu Y, Qiao J. Kisspeptin/Kisspeptin receptor system in the ovary. Front Endocrinol (Lausanne). 2017;8:365.PubMedCrossRef
8.
go back to reference Franssen D, Tena-Sempere M. The kisspeptin receptor: a key G-protein-coupled receptor in the control of the reproductive axis. Best Pract Res Clin Endocrinol Metab. 2018;32(2):107–23.PubMedCrossRef Franssen D, Tena-Sempere M. The kisspeptin receptor: a key G-protein-coupled receptor in the control of the reproductive axis. Best Pract Res Clin Endocrinol Metab. 2018;32(2):107–23.PubMedCrossRef
9.
go back to reference Hu KL, Chang HM, Zhao HC, Yu Y, Li R, Qiao J. Potential roles for the kisspeptin/kisspeptin receptor system in implantation and placentation. Hum Reprod Update. 2019;25(3):326–43.PubMedPubMedCentralCrossRef Hu KL, Chang HM, Zhao HC, Yu Y, Li R, Qiao J. Potential roles for the kisspeptin/kisspeptin receptor system in implantation and placentation. Hum Reprod Update. 2019;25(3):326–43.PubMedPubMedCentralCrossRef
10.
go back to reference Tsutsui K, Bentley GE, Kriegsfeld LJ, Osugi T, Seong JY, Vaudry H. Discovery and evolutionary history of gonadotrophin-inhibitory hormone and kisspeptin: new key neuropeptides controlling reproduction. J Neuroendocrinol. 2010;22(7):716–27.PubMedPubMedCentral Tsutsui K, Bentley GE, Kriegsfeld LJ, Osugi T, Seong JY, Vaudry H. Discovery and evolutionary history of gonadotrophin-inhibitory hormone and kisspeptin: new key neuropeptides controlling reproduction. J Neuroendocrinol. 2010;22(7):716–27.PubMedPubMedCentral
11.
go back to reference Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman BE, et al. KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J Natl Cancer Inst. 1996;88(23):1731–7.PubMedCrossRef Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman BE, et al. KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J Natl Cancer Inst. 1996;88(23):1731–7.PubMedCrossRef
12.
go back to reference Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E, et al. The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem. 2001;276(37):34631–6.PubMedCrossRef Kotani M, Detheux M, Vandenbogaerde A, Communi D, Vanderwinden JM, Le Poul E, et al. The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem. 2001;276(37):34631–6.PubMedCrossRef
13.
go back to reference Marraudino M, Martini M, Trova S, Farinetti A, Ponti G, Gotti S, et al. Kisspeptin system in ovariectomized mice: estradiol and progesterone regulation. Brain Res. 2018;1688:8–14.PubMedCrossRef Marraudino M, Martini M, Trova S, Farinetti A, Ponti G, Gotti S, et al. Kisspeptin system in ovariectomized mice: estradiol and progesterone regulation. Brain Res. 2018;1688:8–14.PubMedCrossRef
14.
go back to reference Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D, Dixon J, et al. Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. Proc Natl Acad Sci U S A. 2005;102(5):1761–6.PubMedPubMedCentralCrossRef Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D, Dixon J, et al. Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. Proc Natl Acad Sci U S A. 2005;102(5):1761–6.PubMedPubMedCentralCrossRef
15.
go back to reference de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100(19):10972–6.PubMedPubMedCentralCrossRef de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci U S A. 2003;100(19):10972–6.PubMedPubMedCentralCrossRef
16.
go back to reference Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS Jr, Shagoury JK, et al. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349(17):1614–27.PubMedCrossRef Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS Jr, Shagoury JK, et al. The GPR54 gene as a regulator of puberty. N Engl J Med. 2003;349(17):1614–27.PubMedCrossRef
17.
go back to reference Uenoyama Y, Nakamura S, Hayakawa Y, Ikegami K, Watanabe Y, Deura C, et al. Lack of pulse and surge modes and glutamatergic stimulation of luteinising hormone release in Kiss1 knockout rats. J Neuroendocrinol. 2015;27(3):187–97.PubMedCrossRef Uenoyama Y, Nakamura S, Hayakawa Y, Ikegami K, Watanabe Y, Deura C, et al. Lack of pulse and surge modes and glutamatergic stimulation of luteinising hormone release in Kiss1 knockout rats. J Neuroendocrinol. 2015;27(3):187–97.PubMedCrossRef
18.
go back to reference Oh YJ, Rhie YJ, Nam HK, Kim HR, Lee KH. Genetic variations of the KISS1R gene in Korean girls with central precocious puberty. J Korean Med Sci. 2017;32(1):108–14.PubMedCrossRef Oh YJ, Rhie YJ, Nam HK, Kim HR, Lee KH. Genetic variations of the KISS1R gene in Korean girls with central precocious puberty. J Korean Med Sci. 2017;32(1):108–14.PubMedCrossRef
19.
go back to reference Silveira LG, Noel SD, Silveira-Neto AP, Abreu AP, Brito VN, Santos MG, et al. Mutations of the KISS1 gene in disorders of puberty. J Clin Endocrinol Metab. 2010;95(5):2276–80.PubMedPubMedCentralCrossRef Silveira LG, Noel SD, Silveira-Neto AP, Abreu AP, Brito VN, Santos MG, et al. Mutations of the KISS1 gene in disorders of puberty. J Clin Endocrinol Metab. 2010;95(5):2276–80.PubMedPubMedCentralCrossRef
20.
go back to reference Shahed A, Young KA. Differential ovarian expression of KiSS-1 and GPR-54 during the estrous cycle and photoperiod induced recrudescence in Siberian hamsters (Phodopus sungorus). Mol Reprod Dev. 2009;76(5):444–52.PubMedPubMedCentralCrossRef Shahed A, Young KA. Differential ovarian expression of KiSS-1 and GPR-54 during the estrous cycle and photoperiod induced recrudescence in Siberian hamsters (Phodopus sungorus). Mol Reprod Dev. 2009;76(5):444–52.PubMedPubMedCentralCrossRef
21.
go back to reference Merhi Z, Thornton K, Bonney E, Cipolla MJ, Charron MJ, Buyuk E. Ovarian kisspeptin expression is related to age and to monocyte chemoattractant protein-1. J Assist Reprod Genet. 2016;33(4):535–43.PubMedPubMedCentralCrossRef Merhi Z, Thornton K, Bonney E, Cipolla MJ, Charron MJ, Buyuk E. Ovarian kisspeptin expression is related to age and to monocyte chemoattractant protein-1. J Assist Reprod Genet. 2016;33(4):535–43.PubMedPubMedCentralCrossRef
22.
go back to reference Castellano JM, Gaytan M, Roa J, Vigo E, Navarro VM, Bellido C, et al. Expression of KiSS-1 in rat ovary: putative local regulator of ovulation? Endocrinology. 2006;147(10):4852–62.PubMedCrossRef Castellano JM, Gaytan M, Roa J, Vigo E, Navarro VM, Bellido C, et al. Expression of KiSS-1 in rat ovary: putative local regulator of ovulation? Endocrinology. 2006;147(10):4852–62.PubMedCrossRef
23.
go back to reference Xiao Y, Ni Y, Huang Y, Wu J, Grossmann R, Zhao R. Effects of kisspeptin-10 on progesterone secretion in cultured chicken ovarian granulosa cells from preovulatory (F1-F3) follicles. Peptides. 2011;32(10):2091–7.PubMedCrossRef Xiao Y, Ni Y, Huang Y, Wu J, Grossmann R, Zhao R. Effects of kisspeptin-10 on progesterone secretion in cultured chicken ovarian granulosa cells from preovulatory (F1-F3) follicles. Peptides. 2011;32(10):2091–7.PubMedCrossRef
24.
go back to reference Tanyapanyachon P, Amelkina O, Chatdarong K. The expression of kisspeptin and its receptor in the domestic cat ovary and uterus in different stages of the ovarian cycle. Theriogenology. 2018;117:40–8.PubMedCrossRef Tanyapanyachon P, Amelkina O, Chatdarong K. The expression of kisspeptin and its receptor in the domestic cat ovary and uterus in different stages of the ovarian cycle. Theriogenology. 2018;117:40–8.PubMedCrossRef
25.
go back to reference Cielesh ME, McGrath BM, Scott CJ, Norman ST, Stephen CP. The localization of kisspeptin and kisspeptin receptor in the canine ovary during different stages of the reproductive cycle. Reprod Domest Anim. 2017;52(Suppl 2):24–8.PubMedCrossRef Cielesh ME, McGrath BM, Scott CJ, Norman ST, Stephen CP. The localization of kisspeptin and kisspeptin receptor in the canine ovary during different stages of the reproductive cycle. Reprod Domest Anim. 2017;52(Suppl 2):24–8.PubMedCrossRef
26.
go back to reference Basini G, Grasselli F, Bussolati S, Ciccimarra R, Maranesi M, Bufalari A, et al. Presence and function of kisspeptin/KISS1R system in swine ovarian follicles. Theriogenology. 2018;115:1–8.PubMedCrossRef Basini G, Grasselli F, Bussolati S, Ciccimarra R, Maranesi M, Bufalari A, et al. Presence and function of kisspeptin/KISS1R system in swine ovarian follicles. Theriogenology. 2018;115:1–8.PubMedCrossRef
27.
go back to reference Gaytan F, Gaytan M, Castellano JM, Romero M, Roa J, Aparicio B, et al. KiSS-1 in the mammalian ovary: distribution of kisspeptin in human and marmoset and alterations in KiSS-1 mRNA levels in a rat model of ovulatory dysfunction. Am J Physiol Endocrinol Metab. 2009;296(3):E520–31.PubMedCrossRef Gaytan F, Gaytan M, Castellano JM, Romero M, Roa J, Aparicio B, et al. KiSS-1 in the mammalian ovary: distribution of kisspeptin in human and marmoset and alterations in KiSS-1 mRNA levels in a rat model of ovulatory dysfunction. Am J Physiol Endocrinol Metab. 2009;296(3):E520–31.PubMedCrossRef
28.
go back to reference Ricu MA, Ramirez VD, Paredes AH, Lara HE. Evidence for a celiac ganglion-ovarian kisspeptin neural network in the rat: intraovarian anti-kisspeptin delays vaginal opening and alters estrous cyclicity. Endocrinology. 2012;153(10):4966–77.PubMedCrossRef Ricu MA, Ramirez VD, Paredes AH, Lara HE. Evidence for a celiac ganglion-ovarian kisspeptin neural network in the rat: intraovarian anti-kisspeptin delays vaginal opening and alters estrous cyclicity. Endocrinology. 2012;153(10):4966–77.PubMedCrossRef
29.
go back to reference Laoharatchatathanin T, Terashima R, Yonezawa T, Kurusu S, Kawaminami M. Augmentation of Metastin/Kisspeptin mRNA expression by the Proestrous luteinizing hormone surge in Granulosa cells of rats: implications for Luteinization. Biol Reprod. 2015;93(1):15.PubMedCrossRef Laoharatchatathanin T, Terashima R, Yonezawa T, Kurusu S, Kawaminami M. Augmentation of Metastin/Kisspeptin mRNA expression by the Proestrous luteinizing hormone surge in Granulosa cells of rats: implications for Luteinization. Biol Reprod. 2015;93(1):15.PubMedCrossRef
30.
go back to reference Peng J, Tang M, Zhang BP, Zhang P, Zhong T, Zong T, et al. Kisspeptin stimulates progesterone secretion via the Erk1/2 mitogen-activated protein kinase signaling pathway in rat luteal cells. Fertil Steril. 2013;99(5):1436–43.e1.PubMedCrossRef Peng J, Tang M, Zhang BP, Zhang P, Zhong T, Zong T, et al. Kisspeptin stimulates progesterone secretion via the Erk1/2 mitogen-activated protein kinase signaling pathway in rat luteal cells. Fertil Steril. 2013;99(5):1436–43.e1.PubMedCrossRef
31.
go back to reference Garcia-Galiano D, van Ingen Schenau D, Leon S, Krajnc-Franken MA, Manfredi-Lozano M, Romero-Ruiz A, et al. Kisspeptin signaling is indispensable for neurokinin B, but not glutamate, stimulation of gonadotropin secretion in mice. Endocrinology. 2012;153(1):316–28.PubMedCrossRef Garcia-Galiano D, van Ingen Schenau D, Leon S, Krajnc-Franken MA, Manfredi-Lozano M, Romero-Ruiz A, et al. Kisspeptin signaling is indispensable for neurokinin B, but not glutamate, stimulation of gonadotropin secretion in mice. Endocrinology. 2012;153(1):316–28.PubMedCrossRef
32.
go back to reference Gaytan F, Garcia-Galiano D, Dorfman MD, Manfredi-Lozano M, Castellano JM, Dissen GA, et al. Kisspeptin receptor haplo-insufficiency causes premature ovarian failure despite preserved gonadotropin secretion. Endocrinology. 2014;155(8):3088–97.PubMedPubMedCentralCrossRef Gaytan F, Garcia-Galiano D, Dorfman MD, Manfredi-Lozano M, Castellano JM, Dissen GA, et al. Kisspeptin receptor haplo-insufficiency causes premature ovarian failure despite preserved gonadotropin secretion. Endocrinology. 2014;155(8):3088–97.PubMedPubMedCentralCrossRef
33.
go back to reference Baarends WM, Uilenbroek JT, Kramer P, Hoogerbrugge JW, van Leeuwen EC, Themmen AP, et al. Anti-mullerian hormone and anti-mullerian hormone type II receptor messenger ribonucleic acid expression in rat ovaries during postnatal development, the estrous cycle, and gonadotropin-induced follicle growth. Endocrinology. 1995;136(11):4951–62.PubMedCrossRef Baarends WM, Uilenbroek JT, Kramer P, Hoogerbrugge JW, van Leeuwen EC, Themmen AP, et al. Anti-mullerian hormone and anti-mullerian hormone type II receptor messenger ribonucleic acid expression in rat ovaries during postnatal development, the estrous cycle, and gonadotropin-induced follicle growth. Endocrinology. 1995;136(11):4951–62.PubMedCrossRef
34.
go back to reference Fernandois D, Na E, Cuevas F, Cruz G, Lara HE, Paredes AH. Kisspeptin is involved in ovarian follicular development during aging in rats. J Endocrinol. 2016;228(3):161–70.PubMedCrossRef Fernandois D, Na E, Cuevas F, Cruz G, Lara HE, Paredes AH. Kisspeptin is involved in ovarian follicular development during aging in rats. J Endocrinol. 2016;228(3):161–70.PubMedCrossRef
35.
go back to reference Saadeldin IM, Koo OJ, Kang JT, Kwon DK, Park SJ, Kim SJ, et al. Paradoxical effects of kisspeptin: it enhances oocyte in vitro maturation but has an adverse impact on hatched blastocysts during in vitro culture. Reprod Fertil Dev. 2012;24(5):656–68.PubMedCrossRef Saadeldin IM, Koo OJ, Kang JT, Kwon DK, Park SJ, Kim SJ, et al. Paradoxical effects of kisspeptin: it enhances oocyte in vitro maturation but has an adverse impact on hatched blastocysts during in vitro culture. Reprod Fertil Dev. 2012;24(5):656–68.PubMedCrossRef
36.
go back to reference Rocha AM, Ding J, Lehman M, Smith GD. Kisspeptin and kisspeptin receptor are expressed in mouse oocytes and participate in meiosis resumption. Fertil Steril. 2012;98(3):S22.CrossRef Rocha AM, Ding J, Lehman M, Smith GD. Kisspeptin and kisspeptin receptor are expressed in mouse oocytes and participate in meiosis resumption. Fertil Steril. 2012;98(3):S22.CrossRef
37.
go back to reference Sirois J, Sayasith K, Brown KA, Stock AE, Bouchard N, Dore M. Cyclooxygenase-2 and its role in ovulation: a 2004 account. Hum Reprod Update. 2004;10(5):373–85.PubMedCrossRef Sirois J, Sayasith K, Brown KA, Stock AE, Bouchard N, Dore M. Cyclooxygenase-2 and its role in ovulation: a 2004 account. Hum Reprod Update. 2004;10(5):373–85.PubMedCrossRef
38.
go back to reference Pallais JC, Bo-Abbas Y, Pitteloud N, Crowley WF Jr, Seminara SB. Neuroendocrine, gonadal, placental, and obstetric phenotypes in patients with IHH and mutations in the G-protein coupled receptor, GPR54. Mol Cell Endocrinol. 2006;254-255:70–7.PubMedCrossRef Pallais JC, Bo-Abbas Y, Pitteloud N, Crowley WF Jr, Seminara SB. Neuroendocrine, gonadal, placental, and obstetric phenotypes in patients with IHH and mutations in the G-protein coupled receptor, GPR54. Mol Cell Endocrinol. 2006;254-255:70–7.PubMedCrossRef
39.
go back to reference Owens LA, Abbara A, Lerner A, O'Floinn S, Christopoulos G, Khanjani S, et al. The direct and indirect effects of kisspeptin-54 on granulosa lutein cell function. Hum Reprod. 2018;33(2):292–302.PubMedCrossRef Owens LA, Abbara A, Lerner A, O'Floinn S, Christopoulos G, Khanjani S, et al. The direct and indirect effects of kisspeptin-54 on granulosa lutein cell function. Hum Reprod. 2018;33(2):292–302.PubMedCrossRef
40.
go back to reference Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet (London, England). 2007;370(9588):685–97.CrossRef Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet (London, England). 2007;370(9588):685–97.CrossRef
41.
go back to reference Gorkem U, Togrul C, Arslan E, Sargin Oruc A, Buyukkayaci Duman N. Is there a role for kisspeptin in pathogenesis of polycystic ovary syndrome? Gynecol Endocrinol. 2018;34(2):157–60.PubMedCrossRef Gorkem U, Togrul C, Arslan E, Sargin Oruc A, Buyukkayaci Duman N. Is there a role for kisspeptin in pathogenesis of polycystic ovary syndrome? Gynecol Endocrinol. 2018;34(2):157–60.PubMedCrossRef
42.
go back to reference Mikkelsen JD, Bentsen AH, Ansel L, Simonneaux V, Juul A. Comparison of the effects of peripherally administered kisspeptins. Regul Pept. 2009;152(1–3):95–100.PubMedCrossRef Mikkelsen JD, Bentsen AH, Ansel L, Simonneaux V, Juul A. Comparison of the effects of peripherally administered kisspeptins. Regul Pept. 2009;152(1–3):95–100.PubMedCrossRef
43.
go back to reference Balasch J, Fabregues F, Carmona F, Casamitjana R, Tena-Sempere M. Ovarian luteinizing hormone priming preceding follicle-stimulating hormone stimulation: clinical and endocrine effects in women with long-term hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2009;94(7):2367–73.PubMedCrossRef Balasch J, Fabregues F, Carmona F, Casamitjana R, Tena-Sempere M. Ovarian luteinizing hormone priming preceding follicle-stimulating hormone stimulation: clinical and endocrine effects in women with long-term hypogonadotropic hypogonadism. J Clin Endocrinol Metab. 2009;94(7):2367–73.PubMedCrossRef
44.
go back to reference Chianese R, Ciaramella V, Fasano S, Pierantoni R, Meccariello R. Kisspeptin regulates steroidogenesis and spermiation in anuran amphibian. Reproduction. 2017;154(4):403–14.PubMedCrossRef Chianese R, Ciaramella V, Fasano S, Pierantoni R, Meccariello R. Kisspeptin regulates steroidogenesis and spermiation in anuran amphibian. Reproduction. 2017;154(4):403–14.PubMedCrossRef
45.
go back to reference Pinto FM, Cejudo-Roman A, Ravina CG, Fernandez-Sanchez M, Martin-Lozano D, Illanes M, et al. Characterization of the kisspeptin system in human spermatozoa. Int J Androl. 2012;35(1):63–73.PubMedCrossRef Pinto FM, Cejudo-Roman A, Ravina CG, Fernandez-Sanchez M, Martin-Lozano D, Illanes M, et al. Characterization of the kisspeptin system in human spermatozoa. Int J Androl. 2012;35(1):63–73.PubMedCrossRef
46.
go back to reference Irfan S, Ehmcke J, Shahab M, Wistuba J, Schlatt S. Immunocytochemical localization of kisspeptin and kisspeptin receptor in the primate testis. J Med Primatol. 2016;45(3):105–11.PubMedCrossRef Irfan S, Ehmcke J, Shahab M, Wistuba J, Schlatt S. Immunocytochemical localization of kisspeptin and kisspeptin receptor in the primate testis. J Med Primatol. 2016;45(3):105–11.PubMedCrossRef
47.
go back to reference Anjum S, Krishna A, Sridaran R, Tsutsui K. Localization of gonadotropin-releasing hormone (GnRH), gonadotropin-inhibitory hormone (GnIH), kisspeptin and GnRH receptor and their possible roles in testicular activities from birth to senescence in mice. J Exp Zool A Ecol Genet Physiol. 2012;317(10):630–44.PubMedPubMedCentral Anjum S, Krishna A, Sridaran R, Tsutsui K. Localization of gonadotropin-releasing hormone (GnRH), gonadotropin-inhibitory hormone (GnIH), kisspeptin and GnRH receptor and their possible roles in testicular activities from birth to senescence in mice. J Exp Zool A Ecol Genet Physiol. 2012;317(10):630–44.PubMedPubMedCentral
48.
go back to reference Meccariello R, Chianese R, Chioccarelli T, Ciaramella V, Fasano S, Pierantoni R, et al. Intra-testicular signals regulate germ cell progression and production of qualitatively mature spermatozoa in vertebrates. Front Endocrinol (Lausanne). 2014;5:69. Meccariello R, Chianese R, Chioccarelli T, Ciaramella V, Fasano S, Pierantoni R, et al. Intra-testicular signals regulate germ cell progression and production of qualitatively mature spermatozoa in vertebrates. Front Endocrinol (Lausanne). 2014;5:69.
49.
go back to reference Harms JF, Welch DR, Miele ME. KISS1 metastasis suppression and emergent pathways. Clin Exp Metastasis. 2003;20(1):11–8.PubMedCrossRef Harms JF, Welch DR, Miele ME. KISS1 metastasis suppression and emergent pathways. Clin Exp Metastasis. 2003;20(1):11–8.PubMedCrossRef
50.
go back to reference Zou P, Wang X, Chen Q, Yang H, Zhou N, Sun L, et al. Kisspeptin protein in seminal plasma is positively associated with semen quality: results from the MARHCS study in Chongqing, China. Biomed Res Int. 2019;2019:5129263.PubMedPubMedCentral Zou P, Wang X, Chen Q, Yang H, Zhou N, Sun L, et al. Kisspeptin protein in seminal plasma is positively associated with semen quality: results from the MARHCS study in Chongqing, China. Biomed Res Int. 2019;2019:5129263.PubMedPubMedCentral
51.
go back to reference Mei H, Walters C, Carter R, Colledge WH. Gpr54−/− mice show more pronounced defects in spermatogenesis than Kiss1−/− mice and improved spermatogenesis with age when exposed to dietary phytoestrogens. Reproduction. 2011;141(3):357–66.PubMedCrossRef Mei H, Walters C, Carter R, Colledge WH. Gpr54−/− mice show more pronounced defects in spermatogenesis than Kiss1−/− mice and improved spermatogenesis with age when exposed to dietary phytoestrogens. Reproduction. 2011;141(3):357–66.PubMedCrossRef
52.
go back to reference Wahab F, Quinton R, Seminara SB. The kisspeptin signaling pathway and its role in human isolated GnRH deficiency. Mol Cell Endocrinol. 2011;346(1–2):29–36.PubMedPubMedCentralCrossRef Wahab F, Quinton R, Seminara SB. The kisspeptin signaling pathway and its role in human isolated GnRH deficiency. Mol Cell Endocrinol. 2011;346(1–2):29–36.PubMedPubMedCentralCrossRef
53.
go back to reference Ayturk N, Firat T, Kukner A, Ozogul C, Tore F, Kandirali IE, et al. The effect of kisspeptin on spermatogenesis and apoptosis in rats. Turk J Med Sci. 2017;47(1):334–42.PubMedCrossRef Ayturk N, Firat T, Kukner A, Ozogul C, Tore F, Kandirali IE, et al. The effect of kisspeptin on spermatogenesis and apoptosis in rats. Turk J Med Sci. 2017;47(1):334–42.PubMedCrossRef
54.
go back to reference Samir H, Nagaoka K, Watanabe G. Effect of kisspeptin antagonist on goat in vitro Leydig cell steroidogenesis. Theriogenology. 2018;121:134–40.PubMedCrossRef Samir H, Nagaoka K, Watanabe G. Effect of kisspeptin antagonist on goat in vitro Leydig cell steroidogenesis. Theriogenology. 2018;121:134–40.PubMedCrossRef
55.
go back to reference Huma T, Ulla F, Hanif F, Rizak JD, Shahab M. Peripheral administration of kisspeptin antagonist does not alter basal plasma testosterone but decreases plasma adiponectin levels in adult male rhesus macaques. Turk J Biol. 2014;38:450–6.CrossRef Huma T, Ulla F, Hanif F, Rizak JD, Shahab M. Peripheral administration of kisspeptin antagonist does not alter basal plasma testosterone but decreases plasma adiponectin levels in adult male rhesus macaques. Turk J Biol. 2014;38:450–6.CrossRef
56.
go back to reference Zhang P, Tang M, Zhong T, Lin Y, Zong T, Zhong C, et al. Expression and function of kisspeptin during mouse decidualization. PLoS One. 2014;9(5):e97647.PubMedPubMedCentralCrossRef Zhang P, Tang M, Zhong T, Lin Y, Zong T, Zhong C, et al. Expression and function of kisspeptin during mouse decidualization. PLoS One. 2014;9(5):e97647.PubMedPubMedCentralCrossRef
57.
go back to reference Calder M, Chan YM, Raj R, Pampillo M, Elbert A, Noonan M, et al. Implantation failure in female Kiss1−/− mice is independent of their hypogonadic state and can be partially rescued by leukemia inhibitory factor. Endocrinology. 2014;155(8):3065–78.PubMedPubMedCentralCrossRef Calder M, Chan YM, Raj R, Pampillo M, Elbert A, Noonan M, et al. Implantation failure in female Kiss1−/− mice is independent of their hypogonadic state and can be partially rescued by leukemia inhibitory factor. Endocrinology. 2014;155(8):3065–78.PubMedPubMedCentralCrossRef
58.
go back to reference Kelleher AM, Milano-Foster J, Behura SK, Spencer TE. Uterine glands coordinate on-time embryo implantation and impact endometrial decidualization for pregnancy success. Nat Commun. 2018;9(1):2435.PubMedPubMedCentralCrossRef Kelleher AM, Milano-Foster J, Behura SK, Spencer TE. Uterine glands coordinate on-time embryo implantation and impact endometrial decidualization for pregnancy success. Nat Commun. 2018;9(1):2435.PubMedPubMedCentralCrossRef
59.
go back to reference Stewart CL, Kaspar P, Brunet LJ, Bhatt H, Gadi I, Kontgen F, et al. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature. 1992;359(6390):76–9.PubMedCrossRef Stewart CL, Kaspar P, Brunet LJ, Bhatt H, Gadi I, Kontgen F, et al. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature. 1992;359(6390):76–9.PubMedCrossRef
60.
go back to reference Pawar S, Laws MJ, Bagchi IC, Bagchi MK. Uterine epithelial estrogen receptor-alpha controls Decidualization via a paracrine mechanism. Mol Endocrinol. 2015;29(9):1362–74.PubMedPubMedCentralCrossRef Pawar S, Laws MJ, Bagchi IC, Bagchi MK. Uterine epithelial estrogen receptor-alpha controls Decidualization via a paracrine mechanism. Mol Endocrinol. 2015;29(9):1362–74.PubMedPubMedCentralCrossRef
61.
go back to reference Chen JR, Cheng JG, Shatzer T, Sewell L, Hernandez L, Stewart CL. Leukemia inhibitory factor can substitute for nidatory estrogen and is essential to inducing a receptive uterus for implantation but is not essential for subsequent embryogenesis. Endocrinology. 2000;141(12):4365–72.PubMedCrossRef Chen JR, Cheng JG, Shatzer T, Sewell L, Hernandez L, Stewart CL. Leukemia inhibitory factor can substitute for nidatory estrogen and is essential to inducing a receptive uterus for implantation but is not essential for subsequent embryogenesis. Endocrinology. 2000;141(12):4365–72.PubMedCrossRef
62.
go back to reference Bilban M, Ghaffari-Tabrizi N, Hintermann E, Bauer S, Molzer S, Zoratti C, et al. Kisspeptin-10, a KiSS-1/metastin-derived decapeptide, is a physiological invasion inhibitor of primary human trophoblasts. J Cell Sci. 2004;117(Pt 8):1319–28.PubMedCrossRef Bilban M, Ghaffari-Tabrizi N, Hintermann E, Bauer S, Molzer S, Zoratti C, et al. Kisspeptin-10, a KiSS-1/metastin-derived decapeptide, is a physiological invasion inhibitor of primary human trophoblasts. J Cell Sci. 2004;117(Pt 8):1319–28.PubMedCrossRef
63.
go back to reference Oride A, Kanasaki H, Mijiddorj T, Sukhbaatar U, Ishihara T, Kyo S. Regulation of kisspeptin and gonadotropin-releasing hormone expression in rat placenta: study using primary cultures of rat placental cells. Reprod Biol Endocrinol. 2015;13:90.PubMedPubMedCentralCrossRef Oride A, Kanasaki H, Mijiddorj T, Sukhbaatar U, Ishihara T, Kyo S. Regulation of kisspeptin and gonadotropin-releasing hormone expression in rat placenta: study using primary cultures of rat placental cells. Reprod Biol Endocrinol. 2015;13:90.PubMedPubMedCentralCrossRef
64.
go back to reference Horikoshi Y, Matsumoto H, Takatsu Y, Ohtaki T, Kitada C, Usuki S, et al. Dramatic elevation of plasma metastin concentrations in human pregnancy: metastin as a novel placenta-derived hormone in humans. J Clin Endocrinol Metab. 2003;88(2):914–9.PubMedCrossRef Horikoshi Y, Matsumoto H, Takatsu Y, Ohtaki T, Kitada C, Usuki S, et al. Dramatic elevation of plasma metastin concentrations in human pregnancy: metastin as a novel placenta-derived hormone in humans. J Clin Endocrinol Metab. 2003;88(2):914–9.PubMedCrossRef
65.
go back to reference Jayasena CN, Abbara A, Izzi-Engbeaya C, Comninos AN, Harvey RA, Gonzalez Maffe J, et al. Reduced levels of plasma kisspeptin during the antenatal booking visit are associated with increased risk of miscarriage. J Clin Endocrinol Metab. 2014;99(12):E2652–60.PubMedPubMedCentralCrossRef Jayasena CN, Abbara A, Izzi-Engbeaya C, Comninos AN, Harvey RA, Gonzalez Maffe J, et al. Reduced levels of plasma kisspeptin during the antenatal booking visit are associated with increased risk of miscarriage. J Clin Endocrinol Metab. 2014;99(12):E2652–60.PubMedPubMedCentralCrossRef
66.
go back to reference Sullivan-Pyke C, Haisenleder DJ, Senapati S, Nicolais O, Eisenberg E, Sammel MD, et al. Kisspeptin as a new serum biomarker to discriminate miscarriage from viable intrauterine pregnancy. Fertil Steril. 2018;109(1):137–41.e2.PubMedPubMedCentralCrossRef Sullivan-Pyke C, Haisenleder DJ, Senapati S, Nicolais O, Eisenberg E, Sammel MD, et al. Kisspeptin as a new serum biomarker to discriminate miscarriage from viable intrauterine pregnancy. Fertil Steril. 2018;109(1):137–41.e2.PubMedPubMedCentralCrossRef
67.
go back to reference Katugampola H, King PJ, Chatterjee S, Meso M, Duncan AJ, Achermann JC, et al. Kisspeptin is a novel regulator of human fetal adrenocortical development and function: a finding with important implications for the human Fetoplacental unit. J Clin Endocrinol Metab. 2017;102(9):3349–59.PubMedPubMedCentralCrossRef Katugampola H, King PJ, Chatterjee S, Meso M, Duncan AJ, Achermann JC, et al. Kisspeptin is a novel regulator of human fetal adrenocortical development and function: a finding with important implications for the human Fetoplacental unit. J Clin Endocrinol Metab. 2017;102(9):3349–59.PubMedPubMedCentralCrossRef
68.
go back to reference Torricelli M, Novembri R, Conti N, De Falco G, De Bonis M, Petraglia F. Correlation with placental kisspeptin in postterm pregnancy and apoptosis. Reprod Sci. 2012;19(10):1133–7.PubMedCrossRef Torricelli M, Novembri R, Conti N, De Falco G, De Bonis M, Petraglia F. Correlation with placental kisspeptin in postterm pregnancy and apoptosis. Reprod Sci. 2012;19(10):1133–7.PubMedCrossRef
69.
go back to reference Brown D, Yu BD, Joza N, Benit P, Meneses J, Firpo M, et al. Loss of Aif function causes cell death in the mouse embryo, but the temporal progression of patterning is normal. Proc Natl Acad Sci U S A. 2006;103(26):9918–23.PubMedPubMedCentralCrossRef Brown D, Yu BD, Joza N, Benit P, Meneses J, Firpo M, et al. Loss of Aif function causes cell death in the mouse embryo, but the temporal progression of patterning is normal. Proc Natl Acad Sci U S A. 2006;103(26):9918–23.PubMedPubMedCentralCrossRef
70.
go back to reference Athapathu H, Jayawardana MA, Senanayaka L. A study of the incidence of apoptosis in the human placental cells in the last weeks of pregnancy. J Obstet Gynaecol. 2003;23(5):515–7.PubMedCrossRef Athapathu H, Jayawardana MA, Senanayaka L. A study of the incidence of apoptosis in the human placental cells in the last weeks of pregnancy. J Obstet Gynaecol. 2003;23(5):515–7.PubMedCrossRef
71.
go back to reference Ishimoto H, Jaffe RB. Development and function of the human fetal adrenal cortex: a key component in the feto-placental unit. Endocr Rev. 2011;32(3):317–55.PubMedCrossRef Ishimoto H, Jaffe RB. Development and function of the human fetal adrenal cortex: a key component in the feto-placental unit. Endocr Rev. 2011;32(3):317–55.PubMedCrossRef
72.
go back to reference Nakamura Y, Aoki S, Xing Y, Sasano H, Rainey WE. Metastin stimulates aldosterone synthesis in human adrenal cells. Reprod Sci. 2007;14(8):836–45.PubMedCrossRef Nakamura Y, Aoki S, Xing Y, Sasano H, Rainey WE. Metastin stimulates aldosterone synthesis in human adrenal cells. Reprod Sci. 2007;14(8):836–45.PubMedCrossRef
73.
go back to reference Yoshioka K, Ohno Y, Horiguchi Y, Ozu C, Namiki K, Tachibana M. Effects of a KiSS-1 peptide, a metastasis suppressor gene, on the invasive ability of renal cell carcinoma cells through a modulation of a matrix metalloproteinase 2 expression. Life Sci. 2008;83(9–10):332–8.PubMedCrossRef Yoshioka K, Ohno Y, Horiguchi Y, Ozu C, Namiki K, Tachibana M. Effects of a KiSS-1 peptide, a metastasis suppressor gene, on the invasive ability of renal cell carcinoma cells through a modulation of a matrix metalloproteinase 2 expression. Life Sci. 2008;83(9–10):332–8.PubMedCrossRef
74.
go back to reference Cohen M, Bischof P. Factors regulating trophoblast invasion. Gynecol Obstet Investig. 2007;64(3):126–30.CrossRef Cohen M, Bischof P. Factors regulating trophoblast invasion. Gynecol Obstet Investig. 2007;64(3):126–30.CrossRef
75.
go back to reference Hiden U, Bilban M, Knofler M, Desoye G. Kisspeptins and the placenta: regulation of trophoblast invasion. Rev Endocr Metab Disord. 2007;8(1):31–9.PubMedCrossRef Hiden U, Bilban M, Knofler M, Desoye G. Kisspeptins and the placenta: regulation of trophoblast invasion. Rev Endocr Metab Disord. 2007;8(1):31–9.PubMedCrossRef
76.
go back to reference Francis VA, Abera AB, Matjila M, Millar RP, Katz AA. Kisspeptin regulation of genes involved in cell invasion and angiogenesis in first trimester human trophoblast cells. PLoS One. 2014;9(6):e99680.PubMedPubMedCentralCrossRef Francis VA, Abera AB, Matjila M, Millar RP, Katz AA. Kisspeptin regulation of genes involved in cell invasion and angiogenesis in first trimester human trophoblast cells. PLoS One. 2014;9(6):e99680.PubMedPubMedCentralCrossRef
77.
go back to reference Roseweir AK, Katz AA, Millar RP. Kisspeptin-10 inhibits cell migration in vitro via a receptor-GSK3 beta-FAK feedback loop in HTR8SVneo cells. Placenta. 2012;33(5):408–15.PubMedCrossRef Roseweir AK, Katz AA, Millar RP. Kisspeptin-10 inhibits cell migration in vitro via a receptor-GSK3 beta-FAK feedback loop in HTR8SVneo cells. Placenta. 2012;33(5):408–15.PubMedCrossRef
78.
go back to reference Ramaesh T, Logie JJ, Roseweir AK, Millar RP, Walker BR, Hadoke PW, et al. Kisspeptin-10 inhibits angiogenesis in human placental vessels ex vivo and endothelial cells in vitro. Endocrinology. 2010;151(12):5927–34.PubMedCrossRef Ramaesh T, Logie JJ, Roseweir AK, Millar RP, Walker BR, Hadoke PW, et al. Kisspeptin-10 inhibits angiogenesis in human placental vessels ex vivo and endothelial cells in vitro. Endocrinology. 2010;151(12):5927–34.PubMedCrossRef
Metadata
Title
Reproductive functions of Kisspeptin/KISS1R Systems in the Periphery
Authors
Yubin Cao
Zeping Li
Wenyu Jiang
Yan Ling
Haibin Kuang
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Reproductive Biology and Endocrinology / Issue 1/2019
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-019-0511-x

Other articles of this Issue 1/2019

Reproductive Biology and Endocrinology 1/2019 Go to the issue