Skip to main content
Erschienen in: Brain Structure and Function 9/2017

14.06.2017 | Original Article

Sexually dimorphic distribution of Prokr2 neurons revealed by the Prokr2-Cre mouse model

verfasst von: Zaid Mohsen, Hosung Sim, David Garcia-Galiano, Xingfa Han, Nicole Bellefontaine, Thomas L. Saunders, Carol F. Elias

Erschienen in: Brain Structure and Function | Ausgabe 9/2017

Einloggen, um Zugang zu erhalten

Abstract

Prokineticin receptor 2 (PROKR2) is predominantly expressed in the mammalian central nervous system. Loss-of-function mutations of PROKR2 in humans are associated with Kallmann syndrome due to the disruption of gonadotropin releasing hormone neuronal migration and deficient olfactory bulb morphogenesis. PROKR2 has been also implicated in the neuroendocrine control of GnRH neurons post-migration and other physiological systems. However, the brain circuitry and mechanisms associated with these actions have been difficult to investigate mainly due to the widespread distribution of Prokr2-expressing cells, and the lack of animal models and molecular tools. Here, we describe the generation, validation and characterization of a new mouse model that expresses Cre recombinase driven by the Prokr2 promoter, using CRISPR-Cas9 technology. Cre expression was visualized using reporter genes, tdTomato and GFP, in males and females. Expression of Cre-induced reporter genes was found in brain sites previously described to express Prokr2, e.g., the paraventricular and the suprachiasmatic nuclei, and the area postrema. The Prokr2-Cre mouse model was further validated by colocalization of Cre-induced GFP and Prokr2 mRNA. No disruption of Prokr2 expression, GnRH neuronal migration or fertility was observed. Comparative analysis of Prokr2-Cre expression in male and female brains revealed a sexually dimorphic distribution confirmed by in situ hybridization. In females, higher Cre activity was found in the medial preoptic area, ventromedial nucleus of the hypothalamus, arcuate nucleus, medial amygdala and lateral parabrachial nucleus. In males, Cre was higher in the amygdalo-hippocampal area. The sexually dimorphic pattern of Prokr2 expression indicates differential roles in reproductive function and, potentially, in other physiological systems.
Literatur
Zurück zum Zitat Abreu AP et al (2008) Loss-of-function mutations in the genes encoding prokineticin-2 or prokineticin receptor-2 cause autosomal recessive Kallmann syndrome. J Clin Endocrinol Metab 93:4113–4118. doi:10.1210/jc.2008-0958 CrossRefPubMed Abreu AP et al (2008) Loss-of-function mutations in the genes encoding prokineticin-2 or prokineticin receptor-2 cause autosomal recessive Kallmann syndrome. J Clin Endocrinol Metab 93:4113–4118. doi:10.​1210/​jc.​2008-0958 CrossRefPubMed
Zurück zum Zitat Allen SJ, Garcia-Galiano D, Borges BC, Burger LL, Boehm U, Elias CF (2016) Leptin receptor null mice with re-expression of LepR in GnRH-R expressing cells display elevated FSH levels but remain in a prepubertal state Am J Physiol Regul Integr Comp Physiol 00529:02015. doi:10.1152/ajpregu.00529.2015 Allen SJ, Garcia-Galiano D, Borges BC, Burger LL, Boehm U, Elias CF (2016) Leptin receptor null mice with re-expression of LepR in GnRH-R expressing cells display elevated FSH levels but remain in a prepubertal state Am J Physiol Regul Integr Comp Physiol 00529:02015. doi:10.​1152/​ajpregu.​00529.​2015
Zurück zum Zitat Becker K, Jechow B (2011) Generation of transgenic mice by pronuclear microinjection. In: Pease S, Saunders TL (eds) Advanced protocols for animal transgenesis: an ISTT manual. Springer, Berlin, pp 99–116CrossRef Becker K, Jechow B (2011) Generation of transgenic mice by pronuclear microinjection. In: Pease S, Saunders TL (eds) Advanced protocols for animal transgenesis: an ISTT manual. Springer, Berlin, pp 99–116CrossRef
Zurück zum Zitat Beltramino C, Taleisnik S (1978) Facilitatory and inhibitory effects of electrochemical stimulation of the amygdala on the release of luteinizing hormone. Brain Res 144:95–107CrossRefPubMed Beltramino C, Taleisnik S (1978) Facilitatory and inhibitory effects of electrochemical stimulation of the amygdala on the release of luteinizing hormone. Brain Res 144:95–107CrossRefPubMed
Zurück zum Zitat Bressler SC, Baum MJ (1996) Sex comparison of neuronal Fos immunoreactivity in the rat vomeronasal projection circuit after chemosensory stimulation. Neuroscience 71:1063–1072CrossRefPubMed Bressler SC, Baum MJ (1996) Sex comparison of neuronal Fos immunoreactivity in the rat vomeronasal projection circuit after chemosensory stimulation. Neuroscience 71:1063–1072CrossRefPubMed
Zurück zum Zitat Brinster RL, Braun RE, Lo D, Avarbock MR, Oram F, Palmiter RD (1989) Targeted correction of a major histocompatibility class II E alpha gene by DNA microinjected into mouse eggs. Proc Natl Acad Sci USA 86:7087–7091CrossRefPubMedPubMedCentral Brinster RL, Braun RE, Lo D, Avarbock MR, Oram F, Palmiter RD (1989) Targeted correction of a major histocompatibility class II E alpha gene by DNA microinjected into mouse eggs. Proc Natl Acad Sci USA 86:7087–7091CrossRefPubMedPubMedCentral
Zurück zum Zitat Coolen LM, Peters HJ, Veening JG (1996) Fos immunoreactivity in the rat brain following consummatory elements of sexual behavior: a sex comparison. Brain Res 738:67–82CrossRefPubMed Coolen LM, Peters HJ, Veening JG (1996) Fos immunoreactivity in the rat brain following consummatory elements of sexual behavior: a sex comparison. Brain Res 738:67–82CrossRefPubMed
Zurück zum Zitat Goodwin EC, Rottman FM (1992) The 3′-flanking sequence of the bovine growth hormone gene contains novel elements required for efficient and accurate polyadenylation. J Biol Chem 267:16330–16334PubMed Goodwin EC, Rottman FM (1992) The 3′-flanking sequence of the bovine growth hormone gene contains novel elements required for efficient and accurate polyadenylation. J Biol Chem 267:16330–16334PubMed
Zurück zum Zitat Hu WP, Li JD, Zhang C, Boehmer L, Siegel JM, Zhou QY (2007) Altered circadian and homeostatic sleep regulation in prokineticin 2-deficient mice. Sleep 30:247–256PubMedPubMedCentral Hu WP, Li JD, Zhang C, Boehmer L, Siegel JM, Zhou QY (2007) Altered circadian and homeostatic sleep regulation in prokineticin 2-deficient mice. Sleep 30:247–256PubMedPubMedCentral
Zurück zum Zitat Kollack-Walker S, Newman SW (1995) Mating and agonistic behavior produce different patterns of Fos immunolabeling in the male Syrian hamster brain. Neuroscience 66:721–736CrossRefPubMed Kollack-Walker S, Newman SW (1995) Mating and agonistic behavior produce different patterns of Fos immunolabeling in the male Syrian hamster brain. Neuroscience 66:721–736CrossRefPubMed
Zurück zum Zitat Lin DC-H, Bullock CM, Ehlert FJ, Chen J-L, Tian H, Zhou Q-Y (2002) Identification and molecular characterization of two closely related G protein-coupled receptors activated by prokineticins/endocrine gland vascular endothelial growth factor. J Biol Chem 277:19276–19280. doi:10.1074/jbc.M202139200 CrossRefPubMed Lin DC-H, Bullock CM, Ehlert FJ, Chen J-L, Tian H, Zhou Q-Y (2002) Identification and molecular characterization of two closely related G protein-coupled receptors activated by prokineticins/endocrine gland vascular endothelial growth factor. J Biol Chem 277:19276–19280. doi:10.​1074/​jbc.​M202139200 CrossRefPubMed
Zurück zum Zitat Meredith M, Westberry JM (2004) Distinctive responses in the medial amygdala to same-species and different-species pheromones. J Neurosci 24:5719–5725CrossRefPubMed Meredith M, Westberry JM (2004) Distinctive responses in the medial amygdala to same-species and different-species pheromones. J Neurosci 24:5719–5725CrossRefPubMed
Zurück zum Zitat Musatov S, Chen W, Pfaff DW, Kaplitt MG, Ogawa S (2006) RNAi-mediated silencing of estrogen receptor alpha in the ventromedial nucleus of hypothalamus abolishes female sexual behaviors. Proc Natl Acad Sci USA 103:10456–10460CrossRefPubMedPubMedCentral Musatov S, Chen W, Pfaff DW, Kaplitt MG, Ogawa S (2006) RNAi-mediated silencing of estrogen receptor alpha in the ventromedial nucleus of hypothalamus abolishes female sexual behaviors. Proc Natl Acad Sci USA 103:10456–10460CrossRefPubMedPubMedCentral
Zurück zum Zitat Rasia-Filho AA, Peres TM, Cubilla-Gutierrez FH, Lucion AB (1991) Effect of estradiol implanted in the corticomedial amygdala on the sexual behavior of castrated male rats. Braz J Med Biol Res 24:1041–1049PubMed Rasia-Filho AA, Peres TM, Cubilla-Gutierrez FH, Lucion AB (1991) Effect of estradiol implanted in the corticomedial amygdala on the sexual behavior of castrated male rats. Braz J Med Biol Res 24:1041–1049PubMed
Zurück zum Zitat Sarfati J, Dode C, Young J (2010) Kallmann syndrome caused by mutations in the PROK2 and PROKR2 genes: pathophysiology and genotype-phenotype correlations. Front Horm Res 39:121–132. doi:10.1159/000312698 CrossRefPubMed Sarfati J, Dode C, Young J (2010) Kallmann syndrome caused by mutations in the PROK2 and PROKR2 genes: pathophysiology and genotype-phenotype correlations. Front Horm Res 39:121–132. doi:10.​1159/​000312698 CrossRefPubMed
Zurück zum Zitat Sawchenko PE, Arias C (1995) Evidence for short-loop feedback effects of ACTH on CRF and vasopressin expression in parvocellular neurosecretory neurons. J Neuroendocrinol 7:721–731CrossRefPubMed Sawchenko PE, Arias C (1995) Evidence for short-loop feedback effects of ACTH on CRF and vasopressin expression in parvocellular neurosecretory neurons. J Neuroendocrinol 7:721–731CrossRefPubMed
Zurück zum Zitat Sawchenko PE, Swanson LW, Vale WW (1984) Corticotropin-releasing factor: co-expression within distinct subsets of oxytocin-, vasopressin-, and neurotensin-immunoreactive neurons in the hypothalamus of the male rat. J Neurosci 4:1118–1129PubMed Sawchenko PE, Swanson LW, Vale WW (1984) Corticotropin-releasing factor: co-expression within distinct subsets of oxytocin-, vasopressin-, and neurotensin-immunoreactive neurons in the hypothalamus of the male rat. J Neurosci 4:1118–1129PubMed
Zurück zum Zitat Sawchenko PE, Brown ER, Chan RK, Ericsson A, Li HY, Roland BL, Kovacs KJ (1996) The paraventricular nucleus of the hypothalamus and the functional neuroanatomy of visceromotor responses to stress. Prog Brain Res 107:201–222CrossRefPubMed Sawchenko PE, Brown ER, Chan RK, Ericsson A, Li HY, Roland BL, Kovacs KJ (1996) The paraventricular nucleus of the hypothalamus and the functional neuroanatomy of visceromotor responses to stress. Prog Brain Res 107:201–222CrossRefPubMed
Zurück zum Zitat Scalia F, Winans SS (1975) The differential projections of the olfactory bulb and accessory olfactory bulb in mammals. J Comp Neurol 161:31–55CrossRefPubMed Scalia F, Winans SS (1975) The differential projections of the olfactory bulb and accessory olfactory bulb in mammals. J Comp Neurol 161:31–55CrossRefPubMed
Zurück zum Zitat Shimshek DR et al (2002) Codon-improved Cre recombinase (iCre) expression in the mouse. Genesis 32:19–26CrossRefPubMed Shimshek DR et al (2002) Codon-improved Cre recombinase (iCre) expression in the mouse. Genesis 32:19–26CrossRefPubMed
Zurück zum Zitat Simerly RB, Chang C, Muramatsu M, Swanson LW (1990) Distribution of androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ hybridization study. J Comp Neurol 294:76–95CrossRefPubMed Simerly RB, Chang C, Muramatsu M, Swanson LW (1990) Distribution of androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ hybridization study. J Comp Neurol 294:76–95CrossRefPubMed
Zurück zum Zitat Sinisi AA et al (2008) Homozygous mutation in the prokineticin-receptor2 gene (Val274Asp) presenting as reversible Kallmann syndrome and persistent oligozoospermia: case report. Hum Reprod 23:2380–2384. doi:10.1093/humrep/den247 CrossRefPubMed Sinisi AA et al (2008) Homozygous mutation in the prokineticin-receptor2 gene (Val274Asp) presenting as reversible Kallmann syndrome and persistent oligozoospermia: case report. Hum Reprod 23:2380–2384. doi:10.​1093/​humrep/​den247 CrossRefPubMed
Zurück zum Zitat Soga T et al (2002) Molecular cloning and characterization of prokineticin receptors. Biochem Biophys Acta 1579:173–179PubMed Soga T et al (2002) Molecular cloning and characterization of prokineticin receptors. Biochem Biophys Acta 1579:173–179PubMed
Zurück zum Zitat Swanson LW, Kuypers HG (1980) The paraventricular nucleus of the hypothalamus: cytoarchitectonic subdivisions and organization of projections to the pituitary, dorsal vagal complex, and spinal cord as demonstrated by retrograde fluorescence double-labeling methods. J Comp Neurol 194:555–570CrossRefPubMed Swanson LW, Kuypers HG (1980) The paraventricular nucleus of the hypothalamus: cytoarchitectonic subdivisions and organization of projections to the pituitary, dorsal vagal complex, and spinal cord as demonstrated by retrograde fluorescence double-labeling methods. J Comp Neurol 194:555–570CrossRefPubMed
Zurück zum Zitat Swanson LW, Sawchenko PE (1980) Paraventricular nucleus: a site for the integration of neuroendocrine and autonomic mechanisms. Neuroendocrinology 31:410–417CrossRefPubMed Swanson LW, Sawchenko PE (1980) Paraventricular nucleus: a site for the integration of neuroendocrine and autonomic mechanisms. Neuroendocrinology 31:410–417CrossRefPubMed
Zurück zum Zitat Veening JG, Coolen LM, de Jong TR, Joosten HW, de Boer SF, Koolhaas JM, Olivier B (2005) Do similar neural systems subserve aggressive and sexual behaviour in male rats? Insights from c-Fos and pharmacological studies. Eur J Pharmacol 526:226–239CrossRefPubMed Veening JG, Coolen LM, de Jong TR, Joosten HW, de Boer SF, Koolhaas JM, Olivier B (2005) Do similar neural systems subserve aggressive and sexual behaviour in male rats? Insights from c-Fos and pharmacological studies. Eur J Pharmacol 526:226–239CrossRefPubMed
Zurück zum Zitat Wierman ME, Kiseljak-Vassiliades K, Tobet S (2011) Gonadotropin-releasing hormone (GnRH) neuron migration: initiation, maintenance and cessation as critical steps to ensure normal reproductive function. Front Neuroendocrinol 32:43–52. doi:10.1016/j.yfrne.2010.07.005 CrossRefPubMed Wierman ME, Kiseljak-Vassiliades K, Tobet S (2011) Gonadotropin-releasing hormone (GnRH) neuron migration: initiation, maintenance and cessation as critical steps to ensure normal reproductive function. Front Neuroendocrinol 32:43–52. doi:10.​1016/​j.​yfrne.​2010.​07.​005 CrossRefPubMed
Zurück zum Zitat Wood RI, Coolen LM (1997) Integration of chemosensory and hormonal cues is essential for sexual behaviour in the male Syrian hamster: role of the medial amygdaloid nucleus. Neuroscience 78:1027–1035CrossRefPubMed Wood RI, Coolen LM (1997) Integration of chemosensory and hormonal cues is essential for sexual behaviour in the male Syrian hamster: role of the medial amygdaloid nucleus. Neuroscience 78:1027–1035CrossRefPubMed
Zurück zum Zitat Wood RI, Newman SW (1995) Integration of chemosensory and hormonal cues is essential for mating in the male Syrian hamster. J Neurosci 15:7261–7269PubMed Wood RI, Newman SW (1995) Integration of chemosensory and hormonal cues is essential for mating in the male Syrian hamster. J Neurosci 15:7261–7269PubMed
Metadaten
Titel
Sexually dimorphic distribution of Prokr2 neurons revealed by the Prokr2-Cre mouse model
verfasst von
Zaid Mohsen
Hosung Sim
David Garcia-Galiano
Xingfa Han
Nicole Bellefontaine
Thomas L. Saunders
Carol F. Elias
Publikationsdatum
14.06.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Brain Structure and Function / Ausgabe 9/2017
Print ISSN: 1863-2653
Elektronische ISSN: 1863-2661
DOI
https://doi.org/10.1007/s00429-017-1456-5

Weitere Artikel der Ausgabe 9/2017

Brain Structure and Function 9/2017 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

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