201
|
Salehi MS, Tamadon A, Jafarzadeh Shirazi MR, Namavar MR, Zamiri MJ. The Role of Arginine-Phenylalanine-Amide-Related Peptides in Mammalian Reproduction. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2015; 9:268-76. [PMID: 26644848 PMCID: PMC4671388 DOI: 10.22074/ijfs.2015.4540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 05/20/2014] [Indexed: 01/23/2023]
Abstract
Until 2000 it was believed that gonadotropin-releasing hormone (GnRH) was the
sole regulator of hypophyseal gonadotropes. In 2000, the discovery of a gonadotropin inhibitory hormone (GnIH) initiated a revolution in the field of reproductive
physiology. Identification of GnIH homologues in mammals, the arginine-phenylalanine-amide (RFamide)-related peptides (RFRPs), indicated a similar function.
Subsequently, further works conducted in various laboratories worldwide have
shown that these neuropeptides inhibit the hypothalamic-hypophyseal axis. This review discusses the role of RFRPs in mammalian reproductive processes.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Amin Tamadon
- Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Reza Namavar
- Histomorphometry and Stereology Research Center, Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Zamiri
- Department of Animal Science, College of Agriculture, Shiraz University, Shiraz, Iran
| |
Collapse
|
202
|
Noroozi A, Jafarzadeh Shirazi MR, Tamadon A, Moghadam A, Niazi A. Increased Litter Size and Suckling Intensity Stimulate mRNA of RFamide-related Peptide in Rats. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2015; 9:380-6. [PMID: 26644862 PMCID: PMC4671385 DOI: 10.22074/ijfs.2015.4554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 06/11/2014] [Indexed: 11/12/2022]
Abstract
Background RFamide-related peptide-3 (RFRP-3) inhibits gonadotropin releasing hormone (GnRH) and luteinizing hormone (LH) secretion in rats. This study evaluates the
effects of litter size and suckling intensity on RFRP mRNA expression in the dorsomedial
hypothalamic nucleus (DMH) of rats. Materials and Methods A total of 32 pregnant and 4 non-lactating ovariectomized
(control group) Sprague-Dawley rats were used in this experimental study. Lactating rats
were allotted to 8 equal groups. In 3 groups, the litter size was adjusted to 5, 10, or 15
pups upon parturition. Dams were allowed to suckle their pups continuously until 8 days
postpartum. In the other 3 groups, the litter size was adjusted to 5 pups following birth.
These pups were separated from the dams for 6 hours on day 8 postpartum, after which
the pups were allowed to suckle for 2.5, 5, or 7.5 minutes prior to killing the dams. In
2 groups, lactating rats with 10 and 15 pups were separated from their pups for 6 hours
on day 8 postpartum. In these groups, the pups were allowed to suckle their dams for 5
minutes before the dams were killed. All rats were killed on day 8 postpartum and the
DMH was removed from each rat. We evaluated RFRP mRNA expression using realtime polymerase chain reaction (PCR). Results The expression of RFRP mRNA in the DMH increased with increased litter size
and suckling intensity compared to the controls. The effect of suckling intensity on the
expression of RFRP mRNA was more pronounced compared to the litter size. Conclusion Increased litter size and suckling intensity stimulated RFRP mRNA expression in the DMH which might contribute to lactation anestrus in rats.
Collapse
Affiliation(s)
- Atefeh Noroozi
- Department of Animal Sciences, College of Agriculture, Shiraz University, Shiraz, Iran
| | | | - Amin Tamadon
- Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Moghadam
- Biotechnology Institute, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Ali Niazi
- Biotechnology Institute, College of Agriculture, Shiraz University, Shiraz, Iran
| |
Collapse
|
203
|
Henningsen JB, Poirel VJ, Mikkelsen JD, Tsutsui K, Simonneaux V, Gauer F. Sex differences in the photoperiodic regulation of RF-Amide related peptide (RFRP) and its receptor GPR147 in the syrian hamster. J Comp Neurol 2015; 524:1825-38. [PMID: 26518222 DOI: 10.1002/cne.23924] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/20/2015] [Accepted: 10/29/2015] [Indexed: 11/10/2022]
Abstract
RF-(Arg-Phe) related peptides (RFRP-1 and -3) are considered to play a role in the seasonal regulation of reproduction; however, the effect of the peptides depends on species and gender. This study aimed at comparing the RFRP system in male and female Syrian hamsters over long and short photoperiods to investigate the neuroanatomical basis of these differential effects. The neuroanatomical distribution of RFRP neurons and fibers, revealed using an antiserum recognizing RFRP-1 and -3, as well as GPR147 mRNA, are similar in male and female Syrian hamsters. RFRP neurons are mainly found in the medial hypothalamus, whereas RFRP projections and GPR147 mRNA are observed in the preoptic area, anteroventral-periventricular nucleus, suprachiasmatic nucleus, paraventricular nucleus, bed nucleus of the stria terminalis, ventromedial hypothalamus, habenular nucleus, and arcuate nucleus. The number of RFRP neurons is higher in females than in males, and in both sexes, the number of RFRP neurons is reduced in short photoperiods. GPR147 mRNA levels are higher in females than in males and are downregulated in short photoperiods, particularly in females. Interestingly, the number of RFRP-positive fibers in the anteroventral-periventricular nucleus is higher only in females adjusted to a short photoperiod. Our results suggest that the RFRP system, which is strongly regulated by photoperiod in both male and female Syrian hamsters, is particularly important in females, with a distinct role in the anteroventral-periventricular nucleus, possibly in the regulation of the preovulatory luteinizing hormone surge via kisspeptin neurons.
Collapse
Affiliation(s)
- Jo B Henningsen
- Institute of Cellular and Integrative Neurosciences, French National Center for Scientific Research, University of Strasbourg, 67084, Strasbourg, France
| | - Vincent-Joseph Poirel
- Institute of Cellular and Integrative Neurosciences, French National Center for Scientific Research, University of Strasbourg, 67084, Strasbourg, France
| | - Jens D Mikkelsen
- Neurobiology Research Unit, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2750, Denmark
| | - Kazuyoshi Tsutsui
- Department of Biology, Waseda University, and Center for Medical Life Science of Waseda University, Tokyo 162-8480, Japan
| | - Valérie Simonneaux
- Institute of Cellular and Integrative Neurosciences, French National Center for Scientific Research, University of Strasbourg, 67084, Strasbourg, France
| | - François Gauer
- Institute of Cellular and Integrative Neurosciences, French National Center for Scientific Research, University of Strasbourg, 67084, Strasbourg, France
| |
Collapse
|
204
|
Min L, Leon S, Li H, Pinilla L, Carroll RS, Tena-Sempere M, Kaiser UB. RF9 Acts as a KISS1R Agonist In Vivo and In Vitro. Endocrinology 2015; 156:4639-48. [PMID: 26418326 PMCID: PMC4655216 DOI: 10.1210/en.2015-1635] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RF9, a reported antagonist of the mammalian gonadotropin-inhibitory hormone receptor, stimulates gonadotropin secretion in mammals. Recent studies have suggested that the stimulatory effect of RF9 on gonadotropin secretion relies on intact kisspeptin receptor (KISS1R) signaling, but the underlying mechanisms remain to be elucidated. Using Chinese Hamster Ovary cells stably transfected with KISS1R, we show that RF9 binds specifically to KISS1R, with a Kd of 1.6 × 10(-5)M, and stimulates an increase in intracellular calcium and inositol phosphate accumulation in a KISS1R-dependent manner, with EC50 values of 3.0 × 10(-6)M and 1.6 × 10(-7)M, respectively. RF9 also stimulated ERK phosphorylation, with a time course similar to that of kisspeptin-10. RFRP-3, the putative endogenous ligand for NPFFR1, did not stimulate inositol phosphate accumulation or pERK, nor did it alter responses to of kisspeptin-10 or RF9. In agreement with these in vitro data, we found that RF9 stimulated a robust LH increase in Npffr1(-/-) mice, similar to that in wild-type littermates, whereas the stimulatory effect of RF9 was markedly reduced in Kiss1r(-/-) and double Kiss1r(-/-)/Npfrr1(-/-) mice. The stimulatory effect of RF9 on LH secretion was restored by the selective rescue of Kiss1r expression in GnRH neurons, in Kiss1r(-/-T) mice. Taken together, our study demonstrates that RF9 acts primarily as a KISS1R agonist, but not as an allosteric modulator, to stimulate LH secretion. Our findings raise questions regarding the utility of RF9 for assessing NPFF1R function and de-emphasize a predominant role of this signaling system in central regulation of reproduction.
Collapse
Affiliation(s)
- Le Min
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Silvia Leon
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Huan Li
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Leonor Pinilla
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Manuel Tena-Sempere
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| |
Collapse
|
205
|
Hypothalamic inhibition of socio-sexual behaviour by increasing neuroestrogen synthesis. Nat Commun 2015; 5:3061. [PMID: 24430094 PMCID: PMC3905723 DOI: 10.1038/ncomms4061] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/03/2013] [Indexed: 01/18/2023] Open
Abstract
Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that inhibits gonadotropin secretion and socio-sexual behaviours. Oestrogen (neuroestrogen) synthesized in the brain from androgen by aromatase regulates male socio-sexual behaviours. Here we show that GnIH directly activates aromatase and increases neuroestrogen synthesis in the preoptic area (POA) and inhibits socio-sexual behaviours of male quail. Aromatase activity and neuroestrogen concentration in the POA are low in the morning when the birds are active, but neuroestrogen synthesis gradually increases until the evening when the birds become inactive. Centrally administered GnIH in the morning increases neuroestrogen synthesis in the POA and decreases socio-sexual behaviours. Centrally administered 17β-oestradiol at higher doses also inhibits socio-sexual behaviours in the morning. These results suggest that GnIH inhibits male socio-sexual behaviours by increasing neuroestrogen synthesis beyond its optimum concentration for the expression of socio-sexual behaviours. This is the first demonstration of any hypothalamic neuropeptide that directly regulates neuroestrogen synthesis.
Collapse
|
206
|
Soga T, Teo CH, Cham KL, Idris MM, Parhar IS. Early-Life Social Isolation Impairs the Gonadotropin-Inhibitory Hormone Neuronal Activity and Serotonergic System in Male Rats. Front Endocrinol (Lausanne) 2015; 6:172. [PMID: 26617573 PMCID: PMC4639717 DOI: 10.3389/fendo.2015.00172] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/26/2015] [Indexed: 12/15/2022] Open
Abstract
Social isolation in early life deregulates the serotonergic system of the brain, compromising reproductive function. Gonadotropin-inhibitory hormone (GnIH) neurons in the dorsomedial hypothalamic nucleus are critical to the inhibitory regulation of gonadotropin-releasing hormone neuronal activity in the brain and release of luteinizing hormone by the pituitary gland. Although GnIH responds to stress, the role of GnIH in social isolation-induced deregulation of the serotonin system and reproductive function remains unclear. We investigated the effect of social isolation in early life on the serotonergic-GnIH neuronal system using enhanced green fluorescent protein (EGFP)-tagged GnIH transgenic rats. Socially isolated rats were observed for anxious and depressive behaviors. Using immunohistochemistry, we examined c-Fos protein expression in EGFP-GnIH neurons in 9-week-old adult male rats after 6 weeks post-weaning isolation or group housing. We also inspected serotonergic fiber juxtapositions in EGFP-GnIH neurons in control and socially isolated male rats. Socially isolated rats exhibited anxious and depressive behaviors. The total number of EGFP-GnIH neurons was the same in control and socially isolated rats, but c-Fos expression in GnIH neurons was significantly reduced in socially isolated rats. Serotonin fiber juxtapositions on EGFP-GnIH neurons were also lower in socially isolated rats. In addition, levels of tryptophan hydroxylase mRNA expression in the dorsal raphe nucleus were significantly attenuated in these rats. These results suggest that social isolation in early-life results in lower serotonin levels, which reduce GnIH neuronal activity and may lead to reproductive failure.
Collapse
Affiliation(s)
- Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Selangor, Malaysia
| | - Chuin Hau Teo
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Selangor, Malaysia
| | - Kai Lin Cham
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Selangor, Malaysia
| | - Marshita Mohd Idris
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Selangor, Malaysia
| | - Ishwar S. Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University, Selangor, Malaysia
| |
Collapse
|
207
|
Helena CV, Toporikova N, Kalil B, Stathopoulos AM, Pogrebna VV, Carolino RO, Anselmo-Franci JA, Bertram R. KNDy Neurons Modulate the Magnitude of the Steroid-Induced Luteinizing Hormone Surges in Ovariectomized Rats. Endocrinology 2015; 156:4200-13. [PMID: 26302111 PMCID: PMC4606747 DOI: 10.1210/en.2015-1070] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Kisspeptin is the most potent stimulator of LH release. There are two kisspeptin neuronal populations in the rodent brain: in the anteroventral periventricular nucleus (AVPV) and in the arcuate nucleus. The arcuate neurons coexpress kisspeptin, neurokinin B, and dynorphin and are called KNDy neurons. Because estradiol increases kisspeptin expression in the AVPV whereas it inhibits KNDy neurons, AVPV and KNDy neurons have been postulated to mediate the positive and negative feedback effects of estradiol on LH secretion, respectively. Yet the role of KNDy neurons during the positive feedback is not clear. In this study, ovariectomized rats were microinjected bilaterally into the arcuate nucleus with a saporin-conjugated neurokinin B receptor agonist for targeted ablation of approximately 70% of KNDy neurons. In oil-treated animals, ablation of KNDy neurons impaired the rise in LH after ovariectomy and kisspeptin content in both populations. In estradiol-treated animals, KNDy ablation did not influence the negative feedback of steroids during the morning. Surprisingly, KNDy ablation increased the steroid-induced LH surges, accompanied by an increase of kisspeptin content in the AVPV. This increase seems to be due to lack of dynorphin input from KNDy neurons to the AVPV as the following: 1) microinjections of a dynorphin antagonist into the AVPV significantly increased the LH surge in estradiol-treated rats, similar to KNDy ablation, and 2) intra-AVPV microinjections of dynorphin in KNDy-ablated rats restored LH surge levels. Our results suggest that KNDy neurons provide inhibition to AVPV kisspeptin neurons through dynorphin and thus regulate the amplitude of the steroid-induced LH surges.
Collapse
Affiliation(s)
- Cleyde V Helena
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Natalia Toporikova
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Bruna Kalil
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Andrea M Stathopoulos
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Veronika V Pogrebna
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Ruither O Carolino
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Janete A Anselmo-Franci
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Richard Bertram
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| |
Collapse
|
208
|
Ferris JK, Tse MT, Hamson DK, Taves MD, Ma C, McGuire N, Arckens L, Bentley GE, Galea LAM, Floresco SB, Soma KK. Neuronal Gonadotrophin-Releasing Hormone (GnRH) and Astrocytic Gonadotrophin Inhibitory Hormone (GnIH) Immunoreactivity in the Adult Rat Hippocampus. J Neuroendocrinol 2015; 27:772-86. [PMID: 26258544 DOI: 10.1111/jne.12307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 01/17/2023]
Abstract
Gonadotrophin-releasing hormone (GnRH) and gonadotrophin inhibitory hormone (GnIH) are neuropeptides secreted by the hypothalamus that regulate reproduction. GnRH receptors are not only present in the anterior pituitary, but also are abundantly expressed in the hippocampus of rats, suggesting that GnRH regulates hippocampal function. GnIH inhibits pituitary gonadotrophin secretion and is also expressed in the hippocampus of a songbird; its role outside of the reproductive axis is not well established. In the present study, we employed immunohistochemistry to examine three forms of GnRH [mammalian GnRH-I (mGnRH-I), chicken GnRH-II (cGnRH-II) and lamprey GnRH-III (lGnRH-III)] and GnIH in the adult rat hippocampus. No mGnRH-I and cGnRH-II+ cell bodies were present in the hippocampus. Sparse mGnRH-I and cGnRH-II+ fibres were present within the CA1 and CA3 fields of the hippocampus, along the hippocampal fissure, and within the hilus of the dentate gyrus. No lGnRH-III was present in the rodent hippocampus. GnIH-immunoreactivity was present in the hippocampus in cell bodies that resembled astrocytes. Males had more GnIH+ cells in the hilus of the dentate gyrus than females. To confirm the GnIH+ cell body phenotype, we performed double-label immunofluorescence against GnIH, glial fibrillary acidic protein (GFAP) and NeuN. Immunofluorescence revealed that all GnIH+ cell bodies in the hippocampus also contained GFAP, a marker of astrocytes. Taken together, these data suggest that GnRH does not reach GnRH receptors in the rat hippocampus primarily via synaptic release. By contrast, GnIH might be synthesised locally in the rat hippocampus by astrocytes. These data shed light on the sites of action and possible functions of GnRH and GnIH outside of the hypothalamic-pituitary-gonadal axis.
Collapse
Affiliation(s)
- J K Ferris
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - M T Tse
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - D K Hamson
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - M D Taves
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - C Ma
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - N McGuire
- Department of Integrative Biology, University of California-Berkeley, Berkeley, CA, USA
| | - L Arckens
- Laboratory of Neuroplasticity and Neuroproteomics, University of Leuven, Leuven, Belgium
| | - G E Bentley
- Department of Integrative Biology, University of California-Berkeley, Berkeley, CA, USA
| | - L A M Galea
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - S B Floresco
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - K K Soma
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
209
|
Srivastava A, Trivedi N, Malik S, Rani S, Kumar V. Molecular basis of photoperiodic control of reproductive cycle in a subtropical songbird, the Indian weaver bird (Ploceus philippinus). Gen Comp Endocrinol 2015; 220:41-5. [PMID: 25172152 DOI: 10.1016/j.ygcen.2014.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/11/2014] [Accepted: 08/17/2014] [Indexed: 11/28/2022]
Abstract
Less is known about genetic basis of photoperiodic regulation of reproductive cycle in subtropical birds. This study measured the expression levels of DIO2, DIO3, GnRH, and GnIH genes in Indian weaver birds subjected to short days (8h light:16h darkness, 8L:16D) and long days (16L:8D) for 48weeks. Whereas small, reproductively inactive testes were maintained under short days, weaver birds underwent testis recrudescence - regression cycle under long days. Relative expression levels of DIO2, DIO3, GnRH and GnIH genes were quantified by the real-time PCR (qPCR) in hypothalamus of birds (n=4) sampled at the beginning of the experiments, and after 10 and 48weeks of short and long day exposures. These sample times represented photosensitive unstimulated (day 0), and under long days the recrudescence (photostimulated, after 10weeks) and regression (photorefractory, after 48weeks) testicular phases. Birds under short days served as controls. The expression pattern of these genes corresponded with testicular phases. High and low GnRH and DIO2 levels were found in birds with large and small testes, respectively. By-and-large the converse was true for GnIH and DIO3 expression levels. Thus, after 10weeks of exposure, there was a significant difference in the mRNA levels between short and long day birds, with small and large testes, respectively. The results also suggest for a possible rapid switching between DIO2 and DIO3 and GnRH and GnIH expressions during testis maturation - regression cycle in Indian weaver birds.
Collapse
Affiliation(s)
| | - Neerja Trivedi
- Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Shalie Malik
- Department of Zoology, University of Lucknow, Lucknow 226007, India.
| | - Sangeeta Rani
- Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Vinod Kumar
- Department of Zoology, University of Delhi, Delhi 110007, India
| |
Collapse
|
210
|
Abstract
The endocrine hypothalamus constitutes those cells which project to the median eminence and secrete neurohormones into the hypophysial portal blood to act on cells of the anterior pituitary gland. The entire endocrine system is controlled by these peptides. In turn, the hypothalamic neuroendocrine cells are regulated by feedback signals from the endocrine glands and other circulating factors. The neuroendocrine cells are found in specific regions of the hypothalamus and are regulated by afferents from higher brain centers. Integrated function is clearly complex and the networks between and amongst the neuroendocrine cells allows fine control to achieve homeostasis. The entry of hormones and other factors into the brain, either via the cerebrospinal fluid or through fenestrated capillaries (in the basal hypothalamus) is important because it influences the extent to which feedback regulation may be imposed. Recent evidence of the passage of factors from the pars tuberalis and the median eminence casts a new layer in our understanding of neuroendocrine regulation. The function of neuroendocrine cells and the means by which pulsatile secretion is achieved is best understood for the close relationship between gonadotropin releasing hormone and luteinizing hormone, which is reviewed in detail. The secretion of other neurohormones is less rigid, so the relationship between hypothalamic secretion and the relevant pituitary hormones is more complex.
Collapse
Affiliation(s)
- I J Clarke
- Monash University, Department of Physiology, Clayton, Australia
| |
Collapse
|
211
|
Thorson JF, Desaulniers AT, Lee C, White BR, Ford JJ, Lents CA. The role of RFamide-related peptide 3 (RFRP3) in regulation of the neuroendocrine reproductive and growth axes of the boar. Anim Reprod Sci 2015; 159:60-5. [DOI: 10.1016/j.anireprosci.2015.05.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 05/08/2015] [Accepted: 05/15/2015] [Indexed: 11/26/2022]
|
212
|
Paullada-Salmerón JA, Cowan M, Aliaga-Guerrero M, Gómez A, Zanuy S, Mañanos E, Muñoz-Cueto JA. LPXRFa peptide system in the European sea bass: A molecular and immunohistochemical approach. J Comp Neurol 2015; 524:176-98. [DOI: 10.1002/cne.23833] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 01/15/2023]
Affiliation(s)
- José A. Paullada-Salmerón
- Department of Biology; Faculty of Environmental and Marine Sciences, University of Cádiz, Marine Campus of International Excellence (CEIMAR) and Agrifood Campus of International Excellence (ceiA3); E-11510 Puerto Real Spain
- INMAR-CACYTMAR Research Institutes, Puerto Real University Campus; E-11510 Puerto Real Spain
| | - Mairi Cowan
- Department of Biology; Faculty of Environmental and Marine Sciences, University of Cádiz, Marine Campus of International Excellence (CEIMAR) and Agrifood Campus of International Excellence (ceiA3); E-11510 Puerto Real Spain
- INMAR-CACYTMAR Research Institutes, Puerto Real University Campus; E-11510 Puerto Real Spain
| | - María Aliaga-Guerrero
- Department of Biology; Faculty of Environmental and Marine Sciences, University of Cádiz, Marine Campus of International Excellence (CEIMAR) and Agrifood Campus of International Excellence (ceiA3); E-11510 Puerto Real Spain
- INMAR-CACYTMAR Research Institutes, Puerto Real University Campus; E-11510 Puerto Real Spain
| | - Ana Gómez
- Institute of Aquaculture of Torre de la Sal, CSIC; Ribera de Cabanes E-12595 Castellón Spain
| | - Silvia Zanuy
- Institute of Aquaculture of Torre de la Sal, CSIC; Ribera de Cabanes E-12595 Castellón Spain
| | - Evaristo Mañanos
- Institute of Aquaculture of Torre de la Sal, CSIC; Ribera de Cabanes E-12595 Castellón Spain
| | - José A. Muñoz-Cueto
- Department of Biology; Faculty of Environmental and Marine Sciences, University of Cádiz, Marine Campus of International Excellence (CEIMAR) and Agrifood Campus of International Excellence (ceiA3); E-11510 Puerto Real Spain
- INMAR-CACYTMAR Research Institutes, Puerto Real University Campus; E-11510 Puerto Real Spain
| |
Collapse
|
213
|
Russo KA, La JL, Stephens SBZ, Poling MC, Padgaonkar NA, Jennings KJ, Piekarski DJ, Kauffman AS, Kriegsfeld LJ. Circadian Control of the Female Reproductive Axis Through Gated Responsiveness of the RFRP-3 System to VIP Signaling. Endocrinology 2015; 156:2608-18. [PMID: 25872006 PMCID: PMC4475714 DOI: 10.1210/en.2014-1762] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Throughout most of the ovulatory cycle, estrogen negative feedback restrains the GnRH neuronal system. Just before ovulation, however, estrogen negative feedback is removed to permit stimulation of the preovulatory GnRH/LH surge (positive feedback) by the circadian clock in the suprachiasmatic nucleus (SCN). The mammalian ortholog of avian gonadotropin-inhibitory hormone, RFamide-related peptide 3 (RFRP-3), participates in the circadian-timed removal of estrogen negative feedback to permit the LH surge. The present study examined the specific neurochemical means by which the SCN controls RFRP-3 activity and explored whether the RFRP-3 system exhibits time-dependent responsiveness to SCN signaling to precisely time the LH surge. We found that RFRP-3 cells in female Syrian hamsters (Mesocricetus auratus) receive close appositions from SCN-derived vasopressin-ergic and vasoactive intestinal peptide (VIP)-ergic terminal fibers. Central VIP administration markedly suppressed RFRP-3 cellular activity in the evening, but not the morning, relative to saline controls, whereas vasopressin was without effect at either time point. Double-label in situ hybridization for Rfrp-3 and the VIP receptors VPAC1 and VPAC2 revealed that the majority of RFRP-3 cells do not coexpress either receptor in Syrian hamsters or mice, suggesting that SCN VIP-ergic signaling inhibits RFRP-3 cells indirectly. The timing of this VIP-mediated disinhibition is further coordinated via temporally gated responsiveness of RFRP-3 cells to circadian signaling. Together, these findings reveal a novel circadian hierarchy of control coordinating the preovulatory LH surge and ovulation.
Collapse
Affiliation(s)
- Kimberly A Russo
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Janet L La
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Shannon B Z Stephens
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Matthew C Poling
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Namita A Padgaonkar
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Kimberly J Jennings
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - David J Piekarski
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Alexander S Kauffman
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Lance J Kriegsfeld
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
214
|
Vaudry H, Leprince J, Chatenet D, Fournier A, Lambert DG, Le Mével JC, Ohlstein EH, Schwertani A, Tostivint H, Vaudry D. International Union of Basic and Clinical Pharmacology. XCII. Urotensin II, urotensin II-related peptide, and their receptor: from structure to function. Pharmacol Rev 2015; 67:214-58. [PMID: 25535277 DOI: 10.1124/pr.114.009480] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Urotensin II (UII) is a cyclic neuropeptide that was first isolated from the urophysis of teleost fish on the basis of its ability to contract the hindgut. Subsequently, UII was characterized in tetrapods including humans. Phylogenetic studies and synteny analysis indicate that UII and its paralogous peptide urotensin II-related peptide (URP) belong to the somatostatin/cortistatin superfamily. In mammals, the UII and URP genes are primarily expressed in cholinergic neurons of the brainstem and spinal cord. UII and URP mRNAs are also present in various organs notably in the cardiovascular, renal, and endocrine systems. UII and URP activate a common G protein-coupled receptor, called UT, that exhibits relatively high sequence identity with somatostatin, opioid, and galanin receptors. The UT gene is widely expressed in the central nervous system (CNS) and in peripheral tissues including the retina, heart, vascular bed, lung, kidney, adrenal medulla, and skeletal muscle. Structure-activity relationship studies and NMR conformational analysis have led to the rational design of a number of peptidic and nonpeptidic UT agonists and antagonists. Consistent with the wide distribution of UT, UII has now been shown to exert a large array of biologic activities, in particular in the CNS, the cardiovascular system, and the kidney. Here, we review the current knowledge concerning the pleiotropic actions of UII and discusses the possible use of antagonists for future therapeutic applications.
Collapse
Affiliation(s)
- Hubert Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jérôme Leprince
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Chatenet
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Alain Fournier
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David G Lambert
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jean-Claude Le Mével
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Eliot H Ohlstein
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Adel Schwertani
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Hervé Tostivint
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| |
Collapse
|
215
|
Jaroslawska J, Chabowska-Kita A, Kaczmarek MM, Kozak LP. Npvf: Hypothalamic Biomarker of Ambient Temperature Independent of Nutritional Status. PLoS Genet 2015; 11:e1005287. [PMID: 26070086 PMCID: PMC4466399 DOI: 10.1371/journal.pgen.1005287] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 05/18/2015] [Indexed: 01/25/2023] Open
Abstract
The mechanism by which mice, exposed to the cold, mobilize endogenous or exogenous fuel sources for heat production is unknown. To address this issue we carried out experiments using 3 models of obesity in mice: C57BL/6J+/+ (wild-type B6) mice with variable susceptibility to obesity in response to being fed a high-fat diet (HFD), B6. Ucp1-/- mice with variable diet-induced obesity (DIO) and a deficiency in brown fat thermogenesis and B6. Lep-/- with defects in thermogenesis, fat mobilization and hyperphagia. Mice were exposed to the cold and monitored for changes in food intake and body composition to determine their energy balance phenotype. Upon cold exposure wild-type B6 and Ucp1-/- mice with diet-induced obesity burned endogenous fat in direct proportion to their fat reserves and changes in food intake were inversely related to fat mass, whereas leptin-deficient and lean wild-type B6 mice fed a chow diet depended on increased food intake to fuel thermogenesis. Analysis of gene expression in the hypothalamus to uncover a central regulatory mechanism revealed suppression of the Npvf gene in a manner that depends on the reduced ambient temperature and degree of exposure to the cold, but not on adiposity, leptin levels, food intake or functional brown fat. Current knowledge does not provide a clear, definite view of central mechanisms controlling energy balance upon cold-activated thermogenesis. Here we show that upon cold exposure lean mice maintain body composition but increase food intake to fuel thermogenesis, whereas cold-exposed mice with DIO utilize endogenous fat stores and then transition to increased food intake as body composition approaches that of the lean controls. Using knockout mice with leptin and Ucp1 gene deficiency our study indicates that the relative energy utilization from food intake and endogenous energy reserves to maintain body temperature during cold exposure is independent of both leptin action and brown fat-linked thermogenesis. Using a combination of genetic and biological approaches, we demonstrate that Npvf gene expression in the hypothalamus is regulated by changes in ambient temperature in a manner independent of the nutritional status of the mouse.
Collapse
Affiliation(s)
- Julia Jaroslawska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | | | - Monika M. Kaczmarek
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Leslie P. Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- * E-mail:
| |
Collapse
|
216
|
Xiang W, Zhang B, Lv F, Ma Y, Chen H, Chen L, Yang F, Wang P, Chu M. The Inhibitory Effects of RFamide-Related Peptide 3 on Luteinizing Hormone Release Involves an Estradiol-Dependent Manner in Prepubertal but Not in Adult Female Mice. Biol Reprod 2015; 93:30. [PMID: 26063871 DOI: 10.1095/biolreprod.115.128777] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/11/2015] [Indexed: 12/22/2022] Open
Abstract
The mammalian gonadotropin-inhibitory hormone (GnIH) ortholog, RFamide-related peptide (RFRP), is considered to act on gonadotropin-releasing hormone (GnRH) neurons and the pituitary to inhibit gonadotropin synthesis and release. However, there is little evidence documenting whether RFamide-related peptide 3 (RFRP-3) plays a primary role in inhibition of the hypothalamo-pituitary-gonadal (HPG) axis prior to the onset of puberty. The present study aimed to understand the functional significance of the neuropeptide on pubertal development. The developmental changes in reproductive-related gene expression at the mRNA level were investigated in the hypothalamus of female mice. The results indicated that RFRP-3 may be an endogenous inhibitory factor for the activation of the HPG axis prior to the onset of puberty. In addition, centrally administered RFRP-3 significantly suppressed plasma luteinizing hormone (LH) levels in prepubertal female mice. Surprisingly, centrally administered RFRP-3 had no effects on plasma LH levels in ovariectomized (OVX) prepubescent female mice. In contrast, RFRP-3 also inhibited plasma LH levels in OVX prepubescent female mice that were treated with 17beta-estradiol replacement. Our study also examined the effects of RFRP-3 on plasma LH release in adult female mice that were ovariectomized at dioestrus, with or without estradiol (E2). Our results showed that the inhibitory effects of RFRP-3 were independent of E2 status. Quantitative real-time PCR and immunohistochemistry analyses showed that RFRP-3 inhibited GnRH expression at both the mRNA and protein levels in the hypothalamus. These data demonstrated that RFRP-3 could effectively suppress pituitary LH release, via the inhibition of GnRH transcription and translation in prepubescent female mice, which is associated with estrogen signaling pathway and developmental stages.
Collapse
Affiliation(s)
- Wei Xiang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Baoyun Zhang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Fenglin Lv
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Yunxia Ma
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Hang Chen
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Long Chen
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Fang Yang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Pingqing Wang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Mingxing Chu
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
217
|
Osugi T, Ubuka T, Tsutsui K. An evolutionary scenario for gonadotrophin-inhibitory hormone in chordates. J Neuroendocrinol 2015; 27:556-66. [PMID: 25494813 DOI: 10.1111/jne.12246] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/03/2014] [Accepted: 12/08/2014] [Indexed: 12/12/2022]
Abstract
In 2000, we discovered a novel hypothalamic neuropeptide that actively inhibits gonadotrophin release in quail and termed it gonadotrophin-inhibitory hormone (GnIH). GnIH peptides have subsequently been identified in most representative species of gnathostomes. They all share a C-terminal LPXRFamide (X = L or Q) motif. GnIH can inhibit gonadotrophin synthesis and release by decreasing the activity of GnRH neuroes, as well as by directly inhibiting pituitary gonadotrophin secretion in birds and mammals. To investigate the evolutionary origin of GnIH and its ancestral function, we identified a GnIH precursor gene encoding GnIHs from the brain of sea lamprey, the most ancient lineage of vertebrates. Lamprey GnIHs possess a C-terminal PQRFamide motif. In vivo administration of one of lamprey GnIHs stimulated the expression of lamprey GnRH in the hypothalamus and gonadotophin β mRNA in the pituitary. Thus, GnIH may have emerged in agnathans as a stimulatory neuropeptide that subsequently diverged to an inhibitory neuropeptide during the course of evolution from basal vertebrates to later-evolved vertebrates, such as birds and mammals. From a structural point of view, pain modulatory neuropeptides, such as neuropeptide FF (NPFF) and neuropeptide AF, share a C-terminal PQRFamide motif. Because agnathans possess both GnIH and NPFF genes, the origin of GnIH and NPFF genes may date back before the emergence of agnathans. More recently, we identified a novel gene encoding RFamide peptides in the amphioxus. Molecular phylogenetic analysis and synteny analysis indicated that this gene is closely related to the genes of GnIH and NPFF of vertebrates. The results suggest that the identified protochordate gene is similar to the common ancestor of GnIH and NPFF genes, indicating that the origin of GnIH and NPFF may date back to the time of the emergence of early chordates. The GnIH and NPFF genes may have diverged by whole-genome duplication during the course of vertebrate evolution.
Collapse
Affiliation(s)
- T Osugi
- Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Tokyo, Japan
| | - T Ubuka
- Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Tokyo, Japan
| | - K Tsutsui
- Department of Biology and Center for Medical Life Science, Waseda University, Shinjuku, Tokyo, Japan
| |
Collapse
|
218
|
Wang Q, Qi X, Guo Y, Li S, Zhang Y, Liu X, Lin H. Molecular identification of GnIH/GnIHR signal and its reproductive function in protogynous hermaphroditic orange-spotted grouper (Epinephelus coioides). Gen Comp Endocrinol 2015; 216:9-23. [PMID: 25943851 DOI: 10.1016/j.ygcen.2015.04.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 04/24/2015] [Accepted: 04/25/2015] [Indexed: 11/21/2022]
Abstract
Gonadotropin-inhibitory hormone (GnIH) and its receptor (GnIHR) play an important role in reproduction regulation in birds, mammals and some teleost species. In protogynous hermaphroditic orange-spotted grouper (Epinephelus coioides), the GnIH/GnIHR signaling pathway and its reproductive function have not been addressed yet. In this study, GnIH and GnIHR in orange-spotted grouper were characterized. gGnIH possessed three putative peptides (gGnIH-I, -II, -III), while gGnIHR showed the characteristics of G protein-coupled receptor and was clustered with GPR147. Functional assays demonstrated that three synthetic gGnIH peptides significantly decreased the forskolin-induced CRE promoter activity, but only gGnIH-I could significantly decrease SRE promoter activity in COS-7 cells transfected with gGnIHR. During the process of ovarian differentiation and development, gGnIH mRNA level in hypothalamus was low at the gonadal primordium stage with gonia, then increased significantly at the early differentiated gonad with primary growth oocytes, while decreased significantly at the developing gonads with cortical-alveolus and vitellogenic stage oocytes. During MT-induced sex reversal, gGnIH mRNA level in hypothalamus increased significantly when the fish completely reversed from female to male. However, gGnIHR mRNA level in pituitary decreased significantly in intersex and completely reversed male fish. Intraperitoneal injection (i.p.) of three gGnIH peptides significantly decreased GnRH1 mRNA levels in hypothalamus, and gGnIH-II significantly inhibited synthesis of LHβ in pituitary. In summary, we firstly identified the GnIH/GnIHR signal in protogynous orange-spotted grouper, which might be involved in the regulation of the reproductive function of sex differentiation, gonadal development and sex reversal via regulating the synthesis of both GnRH and GtH.
Collapse
Affiliation(s)
- Qingqing Wang
- State Key Laboratory of Biocontrol, The Guangdong Province Key Laboratory for Aquatic Economic Animals, and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xin Qi
- State Key Laboratory of Biocontrol, The Guangdong Province Key Laboratory for Aquatic Economic Animals, and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yin Guo
- State Key Laboratory of Biocontrol, The Guangdong Province Key Laboratory for Aquatic Economic Animals, and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Shuisheng Li
- State Key Laboratory of Biocontrol, The Guangdong Province Key Laboratory for Aquatic Economic Animals, and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yong Zhang
- State Key Laboratory of Biocontrol, The Guangdong Province Key Laboratory for Aquatic Economic Animals, and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, The Guangdong Province Key Laboratory for Aquatic Economic Animals, and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, The Guangdong Province Key Laboratory for Aquatic Economic Animals, and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
219
|
Wahab F, Shahab M, Behr R. The involvement of gonadotropin inhibitory hormone and kisspeptin in the metabolic regulation of reproduction. J Endocrinol 2015; 225:R49-66. [PMID: 25957191 DOI: 10.1530/joe-14-0688] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, kisspeptin (KP) and gonadotropin inhibitory hormone (GnIH), two counteracting neuropeptides, have been acknowledged as significant regulators of reproductive function. KP stimulates reproduction while GnIH inhibits it. These two neuropeptides seem to be pivotal for the modulation of reproductive activity in response to internal and external cues. It is well-documented that the current metabolic status of the body is closely linked to its reproductive output. However, how reproductive function is regulated by the body's energy status is less clear. Recent studies have suggested an active participation of hypothalamic KP and GnIH in the modulation of reproductive function according to available metabolic cues. Expression of KISS1, the KP encoding gene, is decreased while expression of RFRP (NPVF), the gene encoding GnIH, is increased in metabolic deficiency conditions. The lower levels of KP, as suggested by a decrease in KISS1 gene mRNA expression, during metabolic deficiency can be corrected by administration of exogenous KP, which leads to an increase in reproductive hormone levels. Likewise, administration of RF9, a GnIH receptor antagonist, can reverse the inhibitory effect of fasting on testosterone in monkeys. Together, it is likely that the integrated function of both these hypothalamic neuropeptides works as a reproductive output regulator in response to a change in metabolic status. In this review, we have summarized literature from nonprimate and primate studies that demonstrate the involvement of KP and GnIH in the metabolic regulation of reproduction.
Collapse
Affiliation(s)
- F Wahab
- Stem Cell Biology Unit Leibniz Institute for Primate Research, German Primate Center, Kellnerweg 4, D-37077 Göttingen, Germany Laboratory of Reproductive Neuroendocrinology Department of Animal Sciences, Faculty of Biological Sciences, Quiad-i-Azam University, Islamabad, Pakistan
| | - M Shahab
- Stem Cell Biology Unit Leibniz Institute for Primate Research, German Primate Center, Kellnerweg 4, D-37077 Göttingen, Germany Laboratory of Reproductive Neuroendocrinology Department of Animal Sciences, Faculty of Biological Sciences, Quiad-i-Azam University, Islamabad, Pakistan
| | - R Behr
- Stem Cell Biology Unit Leibniz Institute for Primate Research, German Primate Center, Kellnerweg 4, D-37077 Göttingen, Germany Laboratory of Reproductive Neuroendocrinology Department of Animal Sciences, Faculty of Biological Sciences, Quiad-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
220
|
Lynn SE, Perfito N, Guardado D, Bentley GE. Food, stress, and circulating testosterone: Cue integration by the testes, not the brain, in male zebra finches (Taeniopygia guttata). Gen Comp Endocrinol 2015; 215:1-9. [PMID: 25849310 DOI: 10.1016/j.ygcen.2015.03.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/09/2015] [Accepted: 03/26/2015] [Indexed: 01/09/2023]
Abstract
Food abundance is closely associated with reproductive readiness in vertebrates. Food scarcity can activate the hypothalamo-pituitary-adrenal axis, decrease sex steroid secretion, and dampen reproductive behavior. However, the mechanisms underlying these transient effects are unclear. Gonadotropin inhibitory hormone (GnIH), a neuropeptide present in the brain and gonads, is also influenced by glucocorticoids and fasting in some species. We investigated whether fasting stress activated the GnIH system in zebra finches (Taeniopygia guttata), with the potential for downstream effects on reproductive physiology and behavior. We fasted or fed males ad libitum for 10h. Fasting increased corticosterone and decreased testosterone in circulation. To assess whether the decrease in testosterone was mediated by changes in the hypothalamus and/or the gonads, we (1) quantified GnRH- and GnIH-positive neurons in the hypothalamus, (2) assessed hypothalamic gene expression for GnRH and GnIH, and (3) examined gene expression for proteins involved in testosterone synthesis in fasted and control birds. No measure of hypothalamic neuropeptides was related to treatment or circulating steroids. However, birds with higher corticosterone had higher testicular GnIH expression and lower testosterone. StAR and LHR expression were lower in the testes of fasted birds than controls. Thus, the decrease in testosterone was not likely mediated by hypothalamic GnIH, but rather by direct actions of fasting and/or corticosterone on the testes, indicating that the testes can integrate and respond to cues of stress directly. Such local inhibition of testosterone synthesis may allow for rapid and reversible changes in physiology and behavior when conditions are inappropriate for breeding.
Collapse
Affiliation(s)
- Sharon E Lynn
- Department of Biology, The College of Wooster, 931 College Mall, Wooster, OH 44619, United States.
| | - Nicole Perfito
- Department of Integrative Biology, The University of California, Berkeley, 3060 Valley Life Sciences Building, Berkeley, CA 94720, United States
| | - Daisy Guardado
- Department of Integrative Biology, The University of California, Berkeley, 3060 Valley Life Sciences Building, Berkeley, CA 94720, United States
| | - George E Bentley
- Department of Integrative Biology, The University of California, Berkeley, 3060 Valley Life Sciences Building, Berkeley, CA 94720, United States
| |
Collapse
|
221
|
Beltramo M, Robert V, Galibert M, Madinier JB, Marceau P, Dardente H, Decourt C, De Roux N, Lomet D, Delmas AF, Caraty A, Aucagne V. Rational design of triazololipopeptides analogs of kisspeptin inducing a long-lasting increase of gonadotropins. J Med Chem 2015; 58:3459-70. [PMID: 25811530 DOI: 10.1021/jm5019675] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
New potent and selective KISS1R agonists were designed using a combination of rational chemical modifications of the endogenous neuropeptide kisspeptin 10 (KP10). Improved resistance to degradation and presumably reduced renal clearance were obtained by introducing a 1,4-disubstituted 1,2,3-triazole as a proteolysis-resistant amide mimic and a serum albumin-binding motif, respectively. These triazololipopeptides are highly potent full agonists of KISS1R and are >100 selective over the closely related NPFF1R. When injected in ewes with a quiescent reproductive system, the best compound of our series induced a much prolonged increase of luteinizing hormone release compared to KP10 and increased follicle-stimulating hormone plasma concentration. Hence, this KISS1R agonist is a new valuable pharmacological tool to explore the potential of KP system in reproduction control. Furthermore, it represents the first step to develop drugs treating reproductive system disorders due to a reduced activity of the hypothalamo-pituitary-gonadal axis such as delayed puberty, hypothalamic amenorrhea, and hypogonadotropic hypogonadism.
Collapse
Affiliation(s)
- Massimiliano Beltramo
- †UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85; CNRS, UMR7247; Université François Rabelais Tours; IFCE), F-37380 Nouzilly, France
| | - Vincent Robert
- †UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85; CNRS, UMR7247; Université François Rabelais Tours; IFCE), F-37380 Nouzilly, France
| | - Mathieu Galibert
- ‡Centre de Biophysique Moléculaire (CNRS UPR4301), Rue Charles Sadron, F-45071 Orléans Cedex 2, France
| | - Jean-Baptiste Madinier
- ‡Centre de Biophysique Moléculaire (CNRS UPR4301), Rue Charles Sadron, F-45071 Orléans Cedex 2, France
| | - Philippe Marceau
- ‡Centre de Biophysique Moléculaire (CNRS UPR4301), Rue Charles Sadron, F-45071 Orléans Cedex 2, France
| | - Hugues Dardente
- †UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85; CNRS, UMR7247; Université François Rabelais Tours; IFCE), F-37380 Nouzilly, France
| | - Caroline Decourt
- †UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85; CNRS, UMR7247; Université François Rabelais Tours; IFCE), F-37380 Nouzilly, France
| | | | - Didier Lomet
- †UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85; CNRS, UMR7247; Université François Rabelais Tours; IFCE), F-37380 Nouzilly, France
| | - Agnès F Delmas
- ‡Centre de Biophysique Moléculaire (CNRS UPR4301), Rue Charles Sadron, F-45071 Orléans Cedex 2, France
| | - Alain Caraty
- †UMR Physiologie de la Reproduction et des Comportements (INRA, UMR85; CNRS, UMR7247; Université François Rabelais Tours; IFCE), F-37380 Nouzilly, France
| | - Vincent Aucagne
- ‡Centre de Biophysique Moléculaire (CNRS UPR4301), Rue Charles Sadron, F-45071 Orléans Cedex 2, France
| |
Collapse
|
222
|
Kriegsfeld LJ, Ubuka T, Bentley GE, Tsutsui K. Seasonal control of gonadotropin-inhibitory hormone (GnIH) in birds and mammals. Front Neuroendocrinol 2015; 37:65-75. [PMID: 25511257 PMCID: PMC4405439 DOI: 10.1016/j.yfrne.2014.12.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/05/2014] [Accepted: 12/07/2014] [Indexed: 12/14/2022]
Abstract
Animals inhabiting temperate and boreal latitudes experience marked seasonal changes in the quality of their environments and maximize reproductive success by phasing breeding activities with the most favorable time of year. Whereas the specific mechanisms driving seasonal changes in reproductive function vary across species, converging lines of evidence suggest gonadotropin-inhibitory hormone (GnIH) serves as a key component of the neuroendocrine circuitry driving seasonal changes in reproduction and sexual motivation in some species. In addition to anticipating environmental change through transduction of photoperiodic information and modifying reproductive state accordingly, GnIH is also positioned to regulate acute changes in reproductive status should unpredictable conditions manifest throughout the year. The present overview summarizes the role of GnIH in avian and mammalian seasonal breeding while considering the similarities and disparities that have emerged from broad investigations across reproductively photoperiodic species.
Collapse
Affiliation(s)
- Lance J Kriegsfeld
- Department of Psychology and Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA 94720-1650, USA.
| | - Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, Center for Medical Life Science of Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - George E Bentley
- Department of Integrative Biology and Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA 94720-3140, USA
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology, Waseda University, Center for Medical Life Science of Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| |
Collapse
|
223
|
Sahin Z, Canpolat S, Ozcan M, Ozgocer T, Kelestimur H. Kisspeptin antagonist prevents RF9-induced reproductive changes in female rats. Reproduction 2015; 149:465-73. [PMID: 25713426 DOI: 10.1530/rep-14-0683] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The aim of this study was to determine the modulatory effects of peptide 234 (p234) (an antagonist of GPR54 receptors) on kisspeptin and RF9 (an RFamide-related peptide antagonist)-induced changes in reproductive functions and energy balance in female rats. Female Sprague-Dawley rats were weaned on postnatal day (pnd) 21. The animals were intracerebroventricularly cannulated under general anesthesia on pnd 23. Groups of female rats were injected with kisspeptin, RF9, p234, kisspeptin plus p234, or RF9 plus p234, daily. The experiments were ended on the day of first diestrus following pnd 60. Kisspeptin or RF9 alone advanced vaginal opening (VO), which was delayed by administration of kisspeptin antagonist alone. In the rats given kisspeptin plus p234 or RF9 plus p234, VO was not different from control rats. Kisspeptin and RF9 elicited significant elevations in circulating LH levels. Coadministrations of kisspeptin or RF9 with p234 decreased LH levels significantly. The use of p234 alone did not cause any significant change in LH secretion. Kisspeptin decreased both food intake and body weight while RF9 decreased only food intake without affecting body weight. The effects of kisspeptin on energy balance were also reversed by central administration of p234. In conclusion, kisspeptin antagonist, p234, modulates the effects of kisspeptin on reproductive functions and energy balance, whereas RF9 seems to exert only its effects on reproductive functions by means of GPR54 signaling in female rats.
Collapse
Affiliation(s)
- Zafer Sahin
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Sinan Canpolat
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Mete Ozcan
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Tuba Ozgocer
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Haluk Kelestimur
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| |
Collapse
|
224
|
Semaan SJ, Kauffman AS. Daily successive changes in reproductive gene expression and neuronal activation in the brains of pubertal female mice. Mol Cell Endocrinol 2015; 401:84-97. [PMID: 25498961 PMCID: PMC4312730 DOI: 10.1016/j.mce.2014.11.025] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/22/2014] [Accepted: 11/18/2014] [Indexed: 01/01/2023]
Abstract
Puberty is governed by the secretion of gonadotropin releasing hormone (GnRH), but the roles and identities of upstream neuropeptides that control and time puberty remain poorly understood. Indeed, how various reproductive neural gene systems change before and during puberty, and in relation to one another, is not well-characterized. We detailed the daily pubertal profile (from postnatal day [PND] 15 to PND 30) of neural Kiss1 (encoding kisspeptin), Kiss1r (kisspeptin receptor), Tac2 (neurokinin B), and Rfrp (RFRP-3, mammalian GnIH) gene expression and day-to-day c-fos induction in each of these cell types in developing female mice. Kiss1 expression in the AVPV/PeN increased substantially over the pubertal transition, reaching adult levels around vaginal opening (PND 27.5), a pubertal marker. However, AVPV/PeN Kiss1 neurons were not highly activated, as measured by c-fos co-expression, at any pubertal age. In the ARC, Kiss1 and Tac2 cell numbers showed moderate increases across the pubertal period, and neuronal activation of Tac2/Kiss1 cells was moderately elevated at all pubertal ages. Additionally, Kiss1r expression specifically in GnRH neurons was already maximal by PND 15 and did not change with puberty. Conversely, both Rfrp expression and Rfrp/c-fos co-expression in the DMN decreased markedly in the early pre-pubertal stage. This robust decrease of the inhibitory RFRP-3 population may diminish inhibition of GnRH neurons during early puberty. Collectively, our data identify the precise timing of important developmental changes - and in some cases, lack thereof - in gene expression and neuronal activation of key reproductive neuropeptides during puberty, with several changes occurring well before vaginal opening.
Collapse
Affiliation(s)
- Sheila J Semaan
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Kauffman
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
225
|
Zheng L, Su J, Fang R, Jin M, Lei Z, Hou Y, Ma Z, Guo T. Developmental changes in the role of gonadotropin-inhibitory hormone (GnIH) and its receptors in the reproductive axis of male Xiaomeishan pigs. Anim Reprod Sci 2015; 154:113-20. [PMID: 25640458 DOI: 10.1016/j.anireprosci.2015.01.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 11/30/2014] [Accepted: 01/14/2015] [Indexed: 11/18/2022]
Abstract
Gonadotropin-inhibitory hormone (GnIH), a key regulator of vertebrate reproduction, was identified in the Japanese quail in 2000, and RFamide-related peptide-3 (RFRP-3) was found to be a mammalian GnIH ortholog. To further determine its role in the reproductive system of male Xiaomeishan pigs, we systematically investigated changes in GnIH and its receptors (GPR147 and GPR74) during the development of the reproductive axis of male pigs. We also investigated the direct effect of RFRP-3 on the synthesis and secretion of testosterone in Leydig cells in vitro. The expression patterns of GnIH in the reproductive axis of male pigs at different stages of development (postnatal 3, 30, 60, 90, and 120D) were studied using semiquantitative RT-PCR and immunohistochemistry. Our results show that hypothalamic, pituitary and testicular levels of GnIH and its receptors mRNA significantly changed on postnatal day 30 and postnatal day 90. The immunoreactivities of the GnIH proteins were mainly localized to the spermatogenic cells, sustentacular cells and interstitial cells of the testis throughout sexual development. It was confirmed that different doses of GnIH/RFRP-3 inhibited the release and synthesis of testosterone, and impacted on the gene expression of 3β-hydroxysteroid dehydrogenase (3β-HSD) and P450, enzymes that play a key role in the synthesis of testosterone. Together, this research provides molecular and morphological data on the regulation of GnIH in the reproductive development of male pigs.
Collapse
Affiliation(s)
- Lucheng Zheng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Juan Su
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Rui Fang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Mengmeng Jin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhihai Lei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yuanlong Hou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhiyu Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Tingting Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
226
|
Geraghty AC, Muroy SE, Zhao S, Bentley GE, Kriegsfeld LJ, Kaufer D. Knockdown of hypothalamic RFRP3 prevents chronic stress-induced infertility and embryo resorption. eLife 2015; 4. [PMID: 25581095 PMCID: PMC4289855 DOI: 10.7554/elife.04316] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/16/2014] [Indexed: 12/01/2022] Open
Abstract
Whereas it is well established that chronic stress induces female reproductive dysfunction, whether stress negatively impacts fertility and fecundity when applied prior to mating and pregnancy has not been explored. In this study, we show that stress that concludes 4 days prior to mating results in persistent and marked reproductive dysfunction, with fewer successful copulation events, fewer pregnancies in those that successfully mated, and increased embryo resorption. Chronic stress exposure led to elevated expression of the hypothalamic inhibitory peptide, RFamide-related peptide-3 (RFRP3), in regularly cycling females. Remarkably, genetic silencing of RFRP3 during stress using an inducible-targeted shRNA completely alleviates stress-induced infertility in female rats, resulting in mating and pregnancy success rates indistinguishable from non-stress controls. We show that chronic stress has long-term effects on pregnancy success, even post-stressor, that are mediated by RFRP3. This points to RFRP3 as a potential clinically relevant single target for stress-induced infertility. DOI:http://dx.doi.org/10.7554/eLife.04316.001 Infertility has become alarmingly common in otherwise healthy women and around 15% of healthy couples younger than 30 years old are unable to conceive within the first year of trying. High-stress levels are known to decrease short-term fertility in humans and other animals, which may serve to prevent pregnancy during times when food or other resources are in short supply. However, it is not clear if exposure to stress has lasting effects on fertility. Previous studies have found that when male rats experience stress, they release a protein called RFRP3. This protein inhibits brain activity, leading to a reduction in the release of reproductive hormones. Geraghty et al. took a closer look at how stress may cause lasting fertility problems in female rats. The researchers exposed female rats to stress by restricting their movements for 3 hr each day over the course of 18 days, which increased the levels of stress hormones in the animals. They allowed the rats to recover for one full reproductive cycle—equivalent to a month in humans—and found that while their stress hormone levels returned to normal, RFRP3 levels in the brain remained high. Even after the recovery period, the females were less likely to mate. Also, the females that did mate were less likely to become pregnant, and the ones that did were more likely to lose some of the embryos. Overall, the level of reproductive success in these rats was only 21%, down from 76% in the control group (who were not exposed to the stress). Next, Geraghty et al. injected a genetically engineered virus into the brain of the stressed rats to switch off the gene that makes RFRP3 during the stress period. This reduced the levels of the RFRP3 protein and restored the mating, pregnancy, and embryo survival rates to the normal levels seen in unstressed rats. These results suggest that increased levels of RFRP3 during stress can have lasting negative effects on fertility. In the future, developing therapies that lower RFRP3 levels may help individuals who experience fertility problems. DOI:http://dx.doi.org/10.7554/eLife.04316.002
Collapse
Affiliation(s)
- Anna C Geraghty
- Department of Integrative Biology, University of California, Berkeley, Berkeley, United States
| | - Sandra E Muroy
- Department of Integrative Biology, University of California, Berkeley, Berkeley, United States
| | - Sheng Zhao
- Department of Psychology, University of California, Berkeley, Berkeley, United States
| | - George E Bentley
- Department of Integrative Biology, University of California, Berkeley, Berkeley, United States
| | - Lance J Kriegsfeld
- Department of Psychology, University of California, Berkeley, Berkeley, United States
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
227
|
Vega MG, Zarek SM, Bhagwat M, Segars JH. Gonadotropin surge-inhibiting/attenuating factors: a review of current evidence, potential applications, and future directions for research. Mol Reprod Dev 2015; 82:2-16. [PMID: 25581424 DOI: 10.1002/mrd.22439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/29/2014] [Indexed: 11/12/2022]
Abstract
Animal studies in the 1980s suggested the existence of an ovarian hormone, termed gonadotropin surge-inhibiting/attenuating factor (GnSIF/AF), that modulates pituitary secretion of luteinizing hormone (LH). Given the importance of identifying regulatory factors of the hypothalamic-pituitary-ovarian axis and the accumulating data suggesting its existence, we conducted a comprehensive literature search using PubMed, Web of Science, Scopus, and Embase to identify articles related to GnSIF/AF. The search generated 161 publications, of which 97 were included in this study. Several attempts have been made to identify and characterize this hormone and several candidates have been identified, but the protein sequences of these putative GnSIF/AF factors differ widely from one study to another. In addition, while the RF-amide RFRP-3 is known foremost as a neuropeptide, some research supports an ovarian origin for this non-steroidal hormone, thereby suggesting a role for RFRP-3 either as a co-modulator of GnSIF/AF or as a gonadotropin-inhibiting factor in the hypothalamus (GnIH). Discovery of the KNDy neurons that modulate GnRH secretion, on the other hand, further encourages the search for substance(s) that modulate their activity and that indirectly affect LH secretion and the hypothalamic-pituitary-ovarian axis. While it has remained an elusive hormone, GnSIF/AF holds many potential applications for contraception, in vitro fertilization, and/or cancer as well as for understanding polycystic ovary syndrome, metabolic diseases, and/or pubertal development. In this review, we rigorously examine the available evidence regarding the existence of GnSIF/AF, previous attempts at its identification, limitations to its discovery, future directions of research, and potential clinical applications.
Collapse
Affiliation(s)
- Mario G Vega
- Department of Obstetrics and Gynecology, St. Luke's-Roosevelt Hospital, New York City, New York
| | | | | | | |
Collapse
|
228
|
Simonneaux V, Bahougne T. A Multi-Oscillatory Circadian System Times Female Reproduction. Front Endocrinol (Lausanne) 2015; 6:157. [PMID: 26539161 PMCID: PMC4611855 DOI: 10.3389/fendo.2015.00157] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/21/2015] [Indexed: 01/14/2023] Open
Abstract
Rhythms in female reproduction are critical to insure that timing of ovulation coincides with oocyte maturation and optimal sexual arousal. This fine tuning of female reproduction involves both the estradiol feedback as an indicator of oocyte maturation, and the master circadian clock of the suprachiasmatic nuclei (SCN) as an indicator of the time of the day. Herein, we are providing an overview of the state of knowledge regarding the differential inhibitory and stimulatory effects of estradiol at different stages of the reproductive axis, and the mechanisms through which the two main neurotransmitters of the SCN, arginine vasopressin, and vasoactive intestinal peptide, convey daily time cues to the reproductive axis. In addition, we will report the most recent findings on the putative functions of peripheral clocks located throughout the reproductive axis [kisspeptin (Kp) neurons, gonadotropin-releasing hormone neurons, gonadotropic cells, the ovary, and the uterus]. This review will point to the critical position of the Kp neurons of the anteroventral periventricular nucleus, which integrate both the stimulatory estradiol signal, and the daily arginine vasopressinergic signal, while displaying a circadian clock. Finally, given the critical role of the light/dark cycle in the synchronization of female reproduction, we will discuss the impact of circadian disruptions observed during shift-work conditions on female reproductive performance and fertility in both animal model and humans.
Collapse
Affiliation(s)
- Valérie Simonneaux
- Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), Strasbourg, France
- *Correspondence: Valérie Simonneaux, Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), 5 rue Blaise Pascal, Strasbourg 67084, France,
| | - Thibault Bahougne
- Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), Strasbourg, France
- Service d’Endocrinologie et Diabète, Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
229
|
Leon S, Tena-Sempere M. Dissecting the Roles of Gonadotropin-Inhibitory Hormone in Mammals: Studies Using Pharmacological Tools and Genetically Modified Mouse Models. Front Endocrinol (Lausanne) 2015; 6:189. [PMID: 26779117 PMCID: PMC4700143 DOI: 10.3389/fendo.2015.00189] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/10/2015] [Indexed: 12/24/2022] Open
Abstract
Reproduction is essential for perpetuation of the species and, hence, is controlled by a sophisticated network of regulatory factors of central and peripheral origin that integrate at the hypothalamic-pituitary-gonadal (HPG) axis. Among the central regulators of reproduction, kisspeptins, as major stimulatory drivers of gonadotropin-releasing hormone (GnRH) neurosecretion, have drawn considerable interest in the last decade. However, the dynamic, if not cyclic (in the female), nature of reproductive function and the potency of kisspeptins and other stimulatory signals of the HPG axis make tenable the existence of counterbalance inhibitory mechanisms, which may keep stimulation at check and would allow adaptation of reproductive maturation and function to different endogenous and environmental conditions. In this context, discovery of the gonadotropin-inhibitory hormone (GnIH) in birds, and its mammalian homolog, RFRP, opened up the exciting possibility that this inhibitory signal might operate centrally to suppress, directly or indirectly, GnRH/gonadotropin secretion, thus reciprocally cooperating with other stimulatory inputs in the dynamic regulation of the reproductive hypothalamic-pituitary unit. After more than 15 years of active research, the role of GnIH/RFRP in the control of the HPG axis has been documented in different species. Yet, important aspects of the physiology of this system, especially regarding its relative importance and actual roles in the control of key facets of reproductive function, remain controversial. In the present work, we aim to provide a critical review of recent developments in this area, with special attention to studies in rodent models, using pharmacological tools and functional genomics. In doing so, we intend to endow the reader with an updated view of what is known (and what is not known) about the physiological role of GnIH/RFRP signaling in the control of mammalian reproduction.
Collapse
Affiliation(s)
- Silvia Leon
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Instituto de Salud Carlos III, CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia (IMIBIC/HURS), Córdoba, Spain
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Instituto de Salud Carlos III, CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia (IMIBIC/HURS), Córdoba, Spain
- FiDiPro Program, Department of Physiology, University of Turku, Turku, Finland
- *Correspondence: Manuel Tena-Sempere,
| |
Collapse
|
230
|
Tsutsui K, Ubuka T, Son YL, Bentley GE, Kriegsfeld LJ. Contribution of GnIH Research to the Progress of Reproductive Neuroendocrinology. Front Endocrinol (Lausanne) 2015; 6:179. [PMID: 26635728 PMCID: PMC4655308 DOI: 10.3389/fendo.2015.00179] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/05/2015] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of gonadotropin-releasing hormone (GnRH) in mammals at the beginning of the 1970s, it was generally accepted that GnRH is the only hypothalamic neuropeptide regulating gonadotropin release in mammals and other vertebrates. In 2000, however, gonadotropin-inhibitory hormone (GnIH), a novel hypothalamic neuropeptide that actively inhibits gonadotropin release, was discovered in quail. Numerous studies over the past decade and a half have demonstrated that GnIH serves as a key player regulating reproduction across vertebrates, acting on the brain and pituitary to modulate reproductive physiology and behavior. In the latter case, recent evidence indicates that GnIH can regulate reproductive behavior through changes in neurosteroid, such as neuroestrogen, biosynthesis in the brain. This review summarizes the discovery of GnIH, and the contributions to GnIH research focused on its mode of action, regulation of biosynthesis, and how these findings advance our understanding of reproductive neuroendocrinology.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo, Japan
- *Correspondence: Kazuyoshi Tsutsui,
| | - Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo, Japan
- Brain Research Institute Monash Sunway of the Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - You Lee Son
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo, Japan
| | - George E. Bentley
- Department of Integrative Biology, Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Lance J. Kriegsfeld
- Department of Psychology, Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA, USA
| |
Collapse
|
231
|
Shahjahan M, Doi H, Ando H. Differential expression patterns of PQRFamide peptide and its two receptor genes in the brain and pituitary of grass puffer during the reproductive cycle. Gen Comp Endocrinol 2015; 210:152-60. [PMID: 25034121 DOI: 10.1016/j.ygcen.2014.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/01/2014] [Accepted: 07/05/2014] [Indexed: 12/19/2022]
Abstract
Pain-modulatory neuropeptides, PQRFamide (PQRFa) peptides, have recently been implicated in the regulation of reproduction in fish. As a first step toward investigating the role of PQRFa peptides on reproductive function in the grass puffer Takifugu niphobles, which is a semilunar spawner, we cloned genes encoding PQRFa peptide precursor (pqrfa) and its two types of receptors (pqrfa-r1 and pqrfa-r2), and examined changes in their expression levels in the brain and pituitary over several months during the reproductive cycle. The grass puffer PQRFa peptide precursor of 126 amino acid residues contains two putative PQRFa peptides, PQRFa-1 and PQRFa-2, which correspond to NPFF and NPAF in other vertebrates, respectively. The grass puffer PQRFa-R1 and PQRFa-R2 consist of 426 and 453 amino acid residues, respectively, and contain distinct characteristics of G-protein coupled receptors. These three genes were exclusively expressed in the brain and pituitary. The expression levels of pqrfa and pqrfa-r1 were significantly increased during the late stage of sexual maturation, but low in the spawning fish just after releasing sperms and eggs. Therefore, the grass puffer PQRFa peptide may have a role in the late stage of sexual maturation before spawning via PQRFa-R1. In contrast, the pqrfa-r2 expression showed maximum levels in the spawning fish and in the post-spawning period. The present results provide fundamental data suggesting that the grass puffer PQRFa peptide may have multiple roles in the control of reproduction that are dependent on the reproductive stages.
Collapse
Affiliation(s)
- Md Shahjahan
- Department of Fisheries Management, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Hiroyuki Doi
- Shimonoseki Marine Science Museum "Kaikyokan", Shimonoseki Academy of Marine Science, Yamaguchi 750-0036, Japan
| | - Hironori Ando
- Sado Marine Biological Station, Faculty of Science, Niigata University, Sado, Niigata 952-2135, Japan.
| |
Collapse
|
232
|
Glanowska KM, Moenter SM. Differential regulation of GnRH secretion in the preoptic area (POA) and the median eminence (ME) in male mice. Endocrinology 2015; 156:231-41. [PMID: 25314270 PMCID: PMC4272400 DOI: 10.1210/en.2014-1458] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
GnRH release in the median eminence (ME) is the central output for control of reproduction. GnRH processes in the preoptic area (POA) also release GnRH. We examined region-specific regulation of GnRH secretion using fast-scan cyclic voltammetry to detect GnRH release in brain slices from adult male mice. Blocking endoplasmic reticulum calcium reuptake to elevate intracellular calcium evokes GnRH release in both the ME and POA. This release is action potential dependent in the ME but not the POA. Locally applied kisspeptin induced GnRH secretion in both the ME and POA. Local blockade of inositol triphospate-mediated calcium release inhibited kisspeptin-induced GnRH release in the ME, but broad blockade was required in the POA. In contrast, kisspeptin-evoked secretion in the POA was blocked by local gonadotropin-inhibitory hormone, but broad gonadotropin-inhibitory hormone application was required in the ME. Although action potentials are required for GnRH release induced by pharmacologically-increased intracellular calcium in the ME and kisspeptin-evoked release requires inositol triphosphate-mediated calcium release, blocking action potentials did not inhibit kisspeptin-induced GnRH release in the ME. Kisspeptin-induced GnRH release was suppressed after blocking both action potentials and plasma membrane Ca(2+) channels. This suggests that kisspeptin action in the ME requires both increased intracellular calcium and influx from the outside of the cell but not action potentials. Local interactions among kisspeptin and GnRH processes in the ME could thus stimulate GnRH release without involving perisomatic regions of GnRH neurons. Coupling between action potential generation and hormone release in GnRH neurons is thus likely physiologically labile and may vary with region.
Collapse
Affiliation(s)
- Katarzyna M Glanowska
- Neuroscience Graduate Program (K.M.G.), University of Virginia, Charlottesville, Virginia 22908; and Departments of Molecular and Integrative Physiology (S.M.M.), Internal Medicine, and Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan 48109
| | | |
Collapse
|
233
|
Poling MC, Kauffman AS. Regulation and Function of RFRP-3 (GnIH) Neurons during Postnatal Development. Front Endocrinol (Lausanne) 2015; 6:150. [PMID: 26441840 PMCID: PMC4585233 DOI: 10.3389/fendo.2015.00150] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/07/2015] [Indexed: 11/17/2022] Open
Abstract
RFamide-related peptide-3 (RFRP-3) [mammalian ortholog to gonadotropin-inhibiting hormone (GnIH)] potently inhibits gonadotropin secretion in mammals. Studies of RFRP-3 immunoreactivity and Rfrp expression (the gene encoding RFRP-3) in mammalian brains have uncovered several possible pathways regulating RFRP-3 neurons, shedding light on their potential role in reproduction and other processes, and pharmacological studies have probed the target sites of RFRP-3 action. Despite this, there is currently no major consensus on RFRP-3's specific endogenous role(s) in reproductive physiology. Here, we discuss the latest evidence relating to RFRP-3 neuron regulation and function during development and sexual maturation, focusing on rodents. We highlight significant changes in RFRP-3 and Rfrp expression, as well as RFRP-3 neuronal activation, during key stages of postnatal and pubertal development and also discuss recent evidence testing the requisite role of RFRP-3 receptors for normal pubertal timing and developmental LH secretion. Interestingly, some findings suggest that endogenous RFRP-3 signaling may not be necessary for the puberty timing, at least in some species, forcing new hypotheses to be generated regarding this peptide's functional significance to sexual maturation and development.
Collapse
Affiliation(s)
- Matthew C. Poling
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alexander S. Kauffman
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, CA, USA
- *Correspondence: Alexander S. Kauffman, Department of Reproductive Medicine, University of California, San Diego, Leichtag Building, Room 3A-15, 9500 Gilman Drive, #0674, La Jolla, CA 92093, USA,
| |
Collapse
|
234
|
Gojska NM, Friedman Z, Belsham DD. Direct regulation of gonadotrophin-releasing hormone (GnRH) transcription by RF-amide-related peptide-3 and kisspeptin in a novel GnRH-secreting cell line, mHypoA-GnRH/GFP. J Neuroendocrinol 2014; 26:888-97. [PMID: 25283492 DOI: 10.1111/jne.12225] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/09/2014] [Accepted: 09/24/2014] [Indexed: 11/30/2022]
Abstract
RF-amide-related peptide-3 [RFRP-3; also often referred to as the mammalian orthologue of the avian gonadotrophin-inhibitory hormone (GnIH)] and kisspeptin have emerged as potent modulators of neuroendocrine function via direct regulation of the reproductive axis in the hypothalamus and pituitary. There are few studies focusing on the direct regulatory effects of RFRP-3 and kisspeptin on gonadotrophin-releasing hormones (GnRH) neurones. We report their effect on GnRH mRNA expression and release in a novel GnRH neuronal cell model, mHypoA-GnRH/GFP, generated from adult-derived GnRH-GFP neurones. The neurones express receptors for both RFRP-3 and kisspeptin, Gpr147 and Gpr54, respectively. Incubation with 100 nm RFRP-3 results in attenuation of GnRH mRNA expression by approximately 60%. Conversely, incubation with 10 nm of Kiss-10 induced GnRH mRNA expression, whereas the combined effect was an overall repression of GnRH mRNA levels. With transcription inhibitors, the repression of GnRH mRNA levels was linked to a transcriptional mechanism but not mRNA stability. No significant changes in GnRH secretion were observed upon RFRP-3 exposure in these neurones. Our findings suggest that the suppressive signalling of RFRP-3 on GnRH transcription may dominate over kisspeptin induction in the mHypoA-GnRH/GFP GnRH neuronal cell model.
Collapse
Affiliation(s)
- N M Gojska
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
235
|
Ubuka T, Tsutsui K. Evolution of gonadotropin-inhibitory hormone receptor and its ligand. Gen Comp Endocrinol 2014; 209:148-61. [PMID: 25220854 DOI: 10.1016/j.ygcen.2014.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/15/2014] [Accepted: 09/04/2014] [Indexed: 12/13/2022]
Abstract
Gonadotropin-inhibitory hormone (GnIH) is a neuropeptide inhibitor of gonadotropin secretion, which was first identified in the Japanese quail hypothalamus. GnIH peptides share a C-terminal LPXRFamide (X=L or Q) motif in most vertebrates. The receptor for GnIH (GnIHR) is the seven-transmembrane G protein-coupled receptor 147 (GPR147) that inhibits cAMP production. GPR147 is also named neuropeptide FF (NPFF) receptor 1 (NPFFR1), because it also binds NPFF that has a C-terminal PQRFamide motif. To understand the evolutionary history of the GnIH system in the animal kingdom, we searched for receptors structurally similar to GnIHR in the genome of six mammals (human, mouse, rat, cattle, cat, and rabbit), five birds (pigeon, chicken, turkey, budgerigar, and zebra finch), one reptile (green anole), one amphibian (Western clawed flog), six fishes (zebrafish, Nile tilapia, Fugu, coelacanth, spotted gar, and lamprey), one hemichordate (acorn worm), one echinoderm (purple sea urchin), one mollusk (California sea hare), seven insects (pea aphid, African malaria mosquito, honey bee, buff-tailed bumblebee, fruit fly, jewel wasp, and red flour beetle), one cnidarian (hydra), and constructed phylogenetic trees by neighbor joining (NJ) and maximum likelihood (ML) methods. A multiple sequence alignment of the receptors showed highly conserved seven-transmembrane domains as well as disulfide bridge sites between the first and second extracellular loops, including the receptor of hydra. Both NJ and ML analyses grouped the receptors of vertebrates into NPFFR1 and NPFFR2 (GPR74), and the receptors of insects into the receptor for SIFamide peptides that share a C-terminal YRKPPFNGSIFamide motif. Although human, quail and zebrafish GnIHR (NPFFR1) were most structurally similar to SIFamide receptor of fruit fly in the Famide peptide (FMRFamide, neuropeptide F, short neuropeptide F, drosulfakinin, myosuppressin, SIFamide) receptor families, the amino acid sequences and the peptide coding regions of GnIH precursors were most similar to FMRFamide precursor of fruit fly in the precursors of Famide peptide families. Chromosome synteny analysis of the precursor genes of human, quail and zebrafish GnIH and fruit fly Famide peptides further identified conserved synteny in vertebrate GnIH and fruit fly FMRFa precursor genes as well as other Famide peptide precursor genes. These results suggest that GnIH and its receptor pair and SIFamide and its receptor pair may have diverged and co-evolved independently in vertebrates and insects, respectively, from their ancestral Famide peptide and its receptor pair, during diversification and evolution of deuterostomian and protostomian species.
Collapse
Affiliation(s)
- Takayoshi Ubuka
- Department of Biology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan
| | - Kazuyoshi Tsutsui
- Department of Biology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan.
| |
Collapse
|
236
|
Pinelli C, Jadhao AG, Biswas SP, Tsutsui K, D''Aniello B. Neuroanatomical Organization of the Brain Gonadotropin-Inhibitory Hormone and Gonadotropin-Releasing Hormone Systems in the Frog Pelophylax esculentus. BRAIN, BEHAVIOR AND EVOLUTION 2014; 85:15-28. [DOI: 10.1159/000368594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 08/28/2014] [Indexed: 11/19/2022]
Abstract
Growing evidence suggests that gonadotropin-inhibitory hormone (GnIH) may play a key role in mediating vertebrate reproduction. GnIH inhibits gonadotropin synthesis and release by decreasing the activity of gonadotropin-releasing hormone (GnRH) neurons as well as by directly regulating gonadotropin secretion from the pituitary. Whereas the presence of GnIH has been widely investigated in various classes of vertebrates, there are very few immunohistochemical reports focusing on GnIH in amphibians. The aim of this study was to assess the presence and neuroanatomical distribution of GnIH-like immunoreactivity in the brain of the anuran amphibian Pelophylax (Rana) esculentus (esculenta) and to explore any potential anatomical relationship with mammalian GnRH-immunoreactive (mGnRH-ir) elements. The GnIH-like immunoreactive (GnIH-ir) system constitutes two distinct subpopulations in the telencephalon and diencephalon, with the highest number of immunoreactive cells located in the preoptic and suprachiasmatic areas. GnIH-ir neurons were also observed in the medial septum, the anterior commissure, the dorsal hypothalamus, the periventricular nucleus of the hypothalamus, and the posterior tuberculum. Scattered GnIH-ir fibers were present in all major subdivisions of the brain but only occasionally in the median eminence. mGnRH-ir neurons were distributed in the mediobasal telencephalon, the medial septal area, and the anterior preoptic area. Double-label immunohistochemistry revealed that the GnRH and GnIH systems coexist and have overlapping distributions at the level of the anterior preoptic area. Some GnIH-ir fibers were in close proximity to mGnRH-ir cell bodies. Our results suggest that both the neuroanatomy and the functional regulation of GnRH release are conserved properties of the hypothalamic GnIH-ir system among vertebrate species.
Collapse
|
237
|
Jørgensen SR, Andersen MD, Overgaard A, Mikkelsen JD. Changes in RFamide-related peptide-1 (RFRP-1)-immunoreactivity during postnatal development and the estrous cycle. Endocrinology 2014; 155:4402-10. [PMID: 25144921 DOI: 10.1210/en.2014-1274] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH is a key player in the hypothalamic control of gonadotropin secretion from the anterior pituitary gland. It has been shown that the mammalian counterpart of the avian gonadotropin inhibitory hormone named RFamide-related peptide (RFRP) is expressed in hypothalamic neurons that innervate and inhibit GnRH neurons. The RFRP precursor is processed into 2 mature peptides, RFRP-1 and RFRP-3. These are characterized by a conserved C-terminal motif RF-NH2 but display highly different N termini. Even though the 2 peptides are equally potent in vitro, little is known about their relative distribution and their distinct roles in vivo. In this study, we raised an antiserum selective for RFRP-1 and defined the distribution of RFRP-1-immunoreactive (ir) neurons in the rat brain. Next, we analyzed the level of RFRP-1-ir during postnatal development in males and females and investigated changes in RFRP-1-ir during the estrous cycle. RFRP-1-ir neurons were distributed along the third ventricle from the caudal part of the medial anterior hypothalamus throughout the medial tuberal hypothalamus and were localized in, but mostly in between, the dorsomedial hypothalamic, ventromedial hypothalamic, and arcuate nuclei. The number of RFRP-1-ir neurons and the density of cellular immunoreactivity were unchanged from juvenile to adulthood in male rats during the postnatal development. However, both parameters were significantly increased in female rats from peripuberty to adulthood, demonstrating prominent gender difference in the developmental control of RFRP-1 expression. The percentage of c-Fos-positive RFRP-1-ir neurons was significantly higher in diestrus as compared with proestrus and estrus. In conclusion, we found that adult females, as compared with males, have significantly more RFRP-1-ir per cell, and these cells are regulated during the estrous cycle.
Collapse
Affiliation(s)
- Sara R Jørgensen
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | | | | | | |
Collapse
|
238
|
Lima CJG, Cardoso SC, Lemos EFL, Zingler E, Capanema C, Menezes LD, Vogado G, Dos Santos BTA, de Moraes OL, Duarte EF, de Brito VN, Latronico AC, Lofrano-Porto A. Mutational analysis of the genes encoding RFamide-related peptide-3, the human orthologue of gonadotrophin-inhibitory hormone, and its receptor (GPR147) in patients with gonadotrophin-releasing hormone-dependent pubertal disorders. J Neuroendocrinol 2014; 26:817-24. [PMID: 25180599 DOI: 10.1111/jne.12207] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 08/24/2014] [Accepted: 08/27/2014] [Indexed: 11/26/2022]
Abstract
RFamide-related peptide-3 (RFRP-3), the orthologue of avian gonadotrophin-inhibitory hormone, and its receptor GPR147 have been recently identified in the human hypothalamus, and their roles in the regulation of reproductive axis has been studied. The present study aimed to investigate whether the presence of variants in the genes encoding human RFRP-3 (NPVF gene) and its receptor, GPR147 (NPFFR1 gene), is associated with the occurrence of gonadotrophin-releasing hormone-dependent pubertal disorders. Seventy-eight patients with idiopathic central precocious puberty (CPP) and 51 with normosmic isolated hypogonadotrophic hypogonadism (nIHH) were investigated. Fifty healthy subjects comprised the control group. The coding sequences of the NPVF and NPFFR1 genes were amplified and sequenced. Odds ratios (OR) were used to estimate the likelihood of CPP or nIHH in the presence of the described polymorphisms. All such polymorphisms have already been registered in the National Center for Biotechnology Information database. A three-nucleotide in frame deletion was identified in the NPVF gene (p.I71_K72), with a smaller proportion in the CPP (5%) compared to the nIHH (15%) group (P = 0.06). This results in the deletion of the isoleucine at position 71, adjacent to lysine at an endoproteolytic cleavage site of the precursor peptide. This polymorphism was associated with a lower risk of CPP (OR = 0.33; 95% confidence interval = 0.08-0.88); interestingly, only two men with nIHH were homozygotes for this variant. A total of five missense polymorphisms were found in the NPFFR1 gene, which encodes GPR147, with similar frequencies among groups and no association with pubertal timing. Our data suggest that RFRP-3/GPR147 may play secondary, modulatory roles on the regulation of pubertal development; a restraining modulatory effect of the NPVF p.I71_K72 variant on the activation of the gonadotrophic axis cannot be ruled out and deserves further investigation.
Collapse
Affiliation(s)
- C J G Lima
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde e Ambulatório de Endocrinologia das Gônadas e Adrenais, Hospital Universitário de Brasília, Universidade de Brasília, Brasília, DF, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Moussavi M, Wlasichuk M, Chang JP, Habibi HR. Seasonal effects of GnIH on basal and GnRH-induced goldfish somatotrope functions. J Endocrinol 2014; 223:191-202. [PMID: 25319842 DOI: 10.1530/joe-14-0441] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To understand how gonadotropin-inhibitory hormone (GnIH) regulates goldfish GH cell functions, we monitored GH release and expression during early, mid-, and/or late gonadal recrudescence. In vivo and in vitro responses to goldfish (g) GnIH were different, indicating direct action at the level of pituitary, as well as interactions with other neuroendocrine factors involved in GH regulation. Injection of gGnIH consistently reduced basal serum GH levels but elevated pituitary gh mRNA levels, indicating potential dissociation of GH release and synthesis. Goldfish GnRH (sGnRH and cGnRHII) injection differentially stimulated serum GH and pituitary gh mRNA levels with some seasonal differences; these responses were reduced by gGnIH. In contrast, in vitro application of gGnIH during 24-h static incubation of goldfish pituitary cells generally elevated basal GH release and attenuated sGnRH-induced changes in gh mRNA, while suppressing basal gh mRNA levels at mid- and late recrudescence but elevating them at early recrudescence. gGnIH attenuated the GH release responses to sGnRH during static incubation at early, but not at mid- and late recrudescence. In cell column perifusion experiments examining short-term GH release, gGnIH reduced the cGnRHII- and sGnRH-stimulated secretion at late recrudescence but inhibited tha action of cGnRHII only during mid-recrudescence. Interestingly, a reduction of basal GH release upon perifusion with gGnIH during late recrudescence was followed by a rebound increase in GH release upon gGnIH removal. These results indicate that gGnIH exerts complex effects on basal and GnRH-stimulated goldfish GH cell functions and can differentially affect GH release and mRNA expression in a seasonal reproductive manner.
Collapse
Affiliation(s)
- M Moussavi
- Department of Biological SciencesUniversity of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada T2N 1N4Department of Biological SciencesUniversity of Alberta, Edmonton, Alberta, Canada T6G 2E9
| | - M Wlasichuk
- Department of Biological SciencesUniversity of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada T2N 1N4Department of Biological SciencesUniversity of Alberta, Edmonton, Alberta, Canada T6G 2E9 Department of Biological SciencesUniversity of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada T2N 1N4Department of Biological SciencesUniversity of Alberta, Edmonton, Alberta, Canada T6G 2E9
| | - J P Chang
- Department of Biological SciencesUniversity of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada T2N 1N4Department of Biological SciencesUniversity of Alberta, Edmonton, Alberta, Canada T6G 2E9 Department of Biological SciencesUniversity of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada T2N 1N4Department of Biological SciencesUniversity of Alberta, Edmonton, Alberta, Canada T6G 2E9
| | - H R Habibi
- Department of Biological SciencesUniversity of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada T2N 1N4Department of Biological SciencesUniversity of Alberta, Edmonton, Alberta, Canada T6G 2E9
| |
Collapse
|
240
|
Biran J, Golan M, Mizrahi N, Ogawa S, Parhar IS, Levavi-Sivan B. LPXRFa, the piscine ortholog of GnIH, and LPXRF receptor positively regulate gonadotropin secretion in Tilapia (Oreochromis niloticus). Endocrinology 2014; 155:4391-401. [PMID: 25144920 DOI: 10.1210/en.2013-2047] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
LPXRFamide (LPXRFa) peptides have been characterized for their ability to inhibit gonadotropin (GTH) release in birds and stimulate growth hormone (GH) release in frogs. However, their involvement in regulating the reproductive hypothalamo-pituitary-gonadal axis in mammals and fish is inconclusive. To study the role of LPXRFa peptides in the regulation of GTH secretion, we cloned tilapia LPXRFa and LPXRF receptor (LPXRF-R). Processing of the tilapia preproLPXRFa liberated three mature LPXRFa peptides that varied in size and post-translational modifications. Phylogenetic analysis of LPXRFa and the closely related RFamide peptide PQRFa showed clear clustering of each peptide sequence with its orthologs from various vertebrates. Signal-transduction analysis of the tilapia LPXRF-R in COS-7 cells showed clear stimulation of CRE-dependent luciferase activity, whereas the human NPFFR1 showed suppression of forskolin-induced CRE-dependent activity in this system. Administration of the tilapia pyroglutaminated LPXRFa-2 peptide to primary cell culture of tilapia pituitaries, or to reproductive female tilapia by ip injection, positively regulated both LH and FSH release in vivo and in vitro. Using double-labeled fluorescent in-situ hybridization and immunofluorescence, βLH cells were found to co-express both tilapia lpxrf and tilapia lpxrf-r mRNA, whereas some of the βFSH cells coexpressed only lpxrf-r mRNA. No coexpression of tilapia lpxrf-r was identified in GH-positive cells. These findings suggest that the LPXRFa system is a potent positive regulator of the reproductive neuroendocrine axis of tilapia.
Collapse
Affiliation(s)
- Jakob Biran
- Department of Animal Sciences (J.B., M.G., N.M., B.L.S.), The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; and Brain Research Institute (S.O., I.S.P.), Monash University, Sunway Campus, Jalan Lagoon Selatan, Bandar Sunway, Selangor 46150, Malaysia
| | | | | | | | | | | |
Collapse
|
241
|
Ubuka T, Tsutsui K. Review: neuroestrogen regulation of socio-sexual behavior of males. Front Neurosci 2014; 8:323. [PMID: 25352775 PMCID: PMC4195287 DOI: 10.3389/fnins.2014.00323] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/25/2014] [Indexed: 11/13/2022] Open
Abstract
It is thought that estrogen (neuroestrogen) synthesized by the action of aromatase in the brain from testosterone activates male socio-sexual behaviors, such as aggression and sexual behavior in birds. We recently found that gonadotropin-inhibitory hormone (GnIH), a hypothalamic neuropeptide, inhibits socio-sexual behaviors of male quail by directly activating aromatase and increasing neuroestrogen synthesis in the preoptic area (POA). The POA is thought to be the most critical site of aromatization and neuroestrogen action for the regulation of socio-sexual behavior of male birds. We concluded that GnIH inhibits socio-sexual behaviors of male quail by increasing neuroestrogen concentration beyond its optimal concentration in the brain for expression of socio-sexual behavior. On the other hand, it has been reported that dopamine and glutamate, which stimulate male socio-sexual behavior in birds and mammals, inhibit the activity of aromatase in the POA. Multiple studies also report that the activity of aromatase or neuroestrogen is negatively correlated with changes in male socio-sexual behavior in fish, birds, and mammals including humans. Here, we review previous studies that investigated the role of neuroestrogen in the regulation of male socio-sexual behavior and reconsider the hypothesis that neuroestrogen activates male socio-sexual behavior in vertebrates. It is considered that basal concentration of neuroestrogen is required for the maintenance of male socio-sexual behavior but higher concentration of neuroestrogen may inhibit male socio-sexual behavior.
Collapse
Affiliation(s)
| | - Kazuyoshi Tsutsui
- Department of Biology and Center for Medical Life Science, Waseda UniversityShinjuku, Tokyo, Japan
| |
Collapse
|
242
|
Anjum S, Krishna A, Tsutsui K. Inhibitory roles of the mammalian GnIH ortholog RFRP3 in testicular activities in adult mice. J Endocrinol 2014; 223:79-91. [PMID: 25232146 DOI: 10.1530/joe-14-0333] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The aim of this study was to evaluate the effects of in vivo and in vitro treatments with RFamide-related peptide 3 (RFRP3), a mammalian gonadotropin-inhibitory hormone ortholog, on testicular activities, i.e. spermatogenesis and steroidogenesis, in mice. Mice were treated in vivo with different doses of RFRP3 (control: 0.02 μg, 0.2 μg, and 2.0 μg/day) for 8 days. For in vitro study, the testes of mice were evaluated with different doses of RFRP3 (control: 1 and 10 ng/ml) with or without LH (control: 10 and 100 ng/ml) for 24 h at 37 °C. RFRP3 treatment produced significant changes in the body mass, circulating steroid level, and testicular activity in mice. RFRP3 treatment also caused dose-dependent histological changes in spermatogenesis, such as decline in germ cell proliferation and survival markers and increase in apoptotic markers in testis. Both in vivo and in vitro studies showed the inhibitory effect of RFRP3 on testosterone synthesis in the testis. RFRP3 inhibited the expression of the receptor for LH (LHCGR), STAR protein, cytochrome P450 side-chain cleavage (CYP11A1) and 3β-hydroxysteroid dehydrogenase in the testis, and testosterone secretion dose dependently. This study also suggested that the inhibitory effect of RFRP3 in the testis may be mediated through local production of GnRH. Thus, RFRP3 inhibits testicular steroidogenesis and spermatogenesis either indirectly through GnRH or by directly influencing germ cell proliferation, survival, and apoptosis.
Collapse
Affiliation(s)
- Shabana Anjum
- Department of ZoologyBanaras Hindu University, Varanasi 221 005, Uttar Pradesh, IndiaDepartment of BiologyWaseda University, Tokyo 162-8480, Japan
| | - Amitabh Krishna
- Department of ZoologyBanaras Hindu University, Varanasi 221 005, Uttar Pradesh, IndiaDepartment of BiologyWaseda University, Tokyo 162-8480, Japan
| | - Kazuyoshi Tsutsui
- Department of ZoologyBanaras Hindu University, Varanasi 221 005, Uttar Pradesh, IndiaDepartment of BiologyWaseda University, Tokyo 162-8480, Japan
| |
Collapse
|
243
|
A new pathway mediating social effects on the endocrine system: female presence acting via norepinephrine release stimulates gonadotropin-inhibitory hormone in the paraventricular nucleus and suppresses luteinizing hormone in quail. J Neurosci 2014; 34:9803-11. [PMID: 25031417 DOI: 10.1523/jneurosci.3706-13.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Rapid effects of social interactions on transient changes in hormonal levels are known in a wide variety of vertebrate taxa, ranging from fish to humans. Although these responses are mediated by the brain, neurochemical pathways that translate social signals into reproductive physiological changes are unclear. In this study, we analyzed how a female presence modifies synthesis and/or release of various neurochemicals, such as monoamines and neuropeptides, in the brain and downstream reproductive hormones in sexually active male Japanese quail. By viewing a female bird, sexually active males rapidly increased norepinephrine (NE) release in the paraventricular nucleus (PVN) of the hypothalamus, in which gonadotropin-inhibitory hormone (GnIH) neuronal cell bodies exist, increased GnIH precursor mRNA expression in the PVN, and decreased luteinizing hormone (LH) concentration in the plasma. GnIH is a hypothalamic neuropeptide that inhibits gonadotropin secretion from the pituitary. It was further shown that GnIH can rapidly suppress LH release after intravenous administration in this study. Centrally administered NE decreased plasma LH concentration in vivo. It was also shown that NE stimulated the release of GnIH from diencephalic tissue blocks in vitro. Fluorescence double-label immunohistochemistry indicated that GnIH neurons received noradrenergic innervations, and immunohistochemistry combined with in situ hybridization have further shown that GnIH neurons expressed α2A-adrenergic receptor mRNA. These results indicate that a female presence increases NE release in the PVN and stimulates GnIH release, resulting in the suppression of LH release in sexually active male quail.
Collapse
|
244
|
Ubuka T, Tsutsui K. Gonadotropin-inhibitory hormone inhibits aggressive behavior of male quail by increasing neuroestrogen synthesis in the brain beyond its optimum concentration. Gen Comp Endocrinol 2014; 205:49-54. [PMID: 24698787 DOI: 10.1016/j.ygcen.2014.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 03/16/2014] [Indexed: 11/28/2022]
Abstract
The action of testosterone on male socio-sexual behaviors, such as aggressive and sexual behaviors, requires its aromatization into estrogen (neuroestrogen) in the brain. Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that inhibits gonadotropin secretion from the pituitary. On the other hand, wide distribution of GnIH-immunoreactive (ir) neuronal fibers in the brain suggested their roles in the regulation of behavior. Our recent studies have shown that GnIH indeed inhibits aggressive and sexual behaviors. Accordingly, we further investigated the effect of GnIH on aromatase activity and estrogen synthesis in the brain. Abundant GnIH-ir neuronal fibers were observed in the vicinity of aromatase-ir cells in the brain, such as in the preoptic area (POA) that is thought to be the most critical site of aromatization and neuroestrogen action for the regulation of socio-sexual behavior. GnIH receptor (GPR147) mRNA was also expressed in aromatase-ir cells in the POA. GnIH stimulated the activity of aromatase and increased neuroestrogen synthesis in the POA through GPR147. The increase in neuroestrogen concentration in the POA was associated with a significant decrease in aggressive behavior. Finally, centrally administered 17β-estradiol at higher doses inhibited aggressive behavior. These findings indicated that GnIH inhibits aggressive behavior by directly activating aromatase and increasing neuroestrogen synthesis in the brain beyond its optimum concentration for the expression of aggressive behavior. This review highlights recent findings of the role of GnIH in the regulation of neuroestrogen synthesis and its possible function in the regulation of socio-sexual behaviors.
Collapse
Affiliation(s)
- Takayoshi Ubuka
- Department of Biology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan; Department of Biology, College of Liberal Arts and Sciences, Tokyo Medical and Dental University, 2-8-30 Kounodai, Ichikawa, Chiba 272-0827, Japan.
| | - Kazuyoshi Tsutsui
- Department of Biology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan
| |
Collapse
|
245
|
Zhao L, Zhong M, Xue HL, Ding JS, Wang S, Xu JH, Chen L, Xu LX. Effect of RFRP-3 on reproduction is sex- and developmental status-dependent in the striped hamster (Cricetulus barabensis). Gene 2014; 547:273-9. [DOI: 10.1016/j.gene.2014.06.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/07/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
|
246
|
Tsutsui K, Ubuka T. Breakthrough in neuroendocrinology by discovering novel neuropeptides and neurosteroids: 1. Discovery of gonadotropin-inhibitory hormone (GnIH) across vertebrates. Gen Comp Endocrinol 2014; 205:4-10. [PMID: 24704563 DOI: 10.1016/j.ygcen.2014.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 02/26/2014] [Accepted: 03/05/2014] [Indexed: 10/25/2022]
Abstract
Bargmann-Scharrer's discovery of "neurosecretion" in the first half of the 20th century has since matured into the scientific discipline of neuroendocrinology. Identification of novel neurohormones, such as neuropeptides and neurosteroids, is essential for the progress of neuroendocrinology. Our studies over the past two decades have significantly broadened the horizons of this field of research by identifying novel neuropeptides and neurosteroids in vertebrates that have opened new lines of scientific investigation in neuroendocrinology. Since the discovery of gonadotropin-releasing hormone (GnRH) in mammals at the beginning of 1970s, it was generally believed that GnRH is the only hypothalamic neuropeptide regulating gonadotropin release in vertebrates. In 2000, however, we discovered a novel hypothalamic neuropeptide that actively inhibits gonadotropin release in quail and termed it gonadotropin-inhibitory hormone (GnIH). It now appears that GnIH is highly conserved across vertebrates, including humans, and serves a number of behavioral and physiological functions other than regulation of reproduction, providing enormous opportunity for investigators from a wide array of disciplines to study this neuropeptide. This review summarizes the discovery of GnIH and its contribution to the progress of neuroendocrinology.
Collapse
Affiliation(s)
- Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo 162-8480, Japan.
| | - Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology and Center for Medical Life Science, Waseda University, Tokyo 162-8480, Japan
| |
Collapse
|
247
|
Osugi T, Ubuka T, Tsutsui K. Review: evolution of GnIH and related peptides structure and function in the chordates. Front Neurosci 2014; 8:255. [PMID: 25177268 PMCID: PMC4133751 DOI: 10.3389/fnins.2014.00255] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/29/2014] [Indexed: 11/13/2022] Open
Abstract
Discovery of gonadotropin-inhibitory hormone (GnIH) in the Japanese quail in 2000 was the first to demonstrate the existence of a hypothalamic neuropeptide inhibiting gonadotropin release. We now know that GnIH regulates reproduction by inhibiting gonadotropin synthesis and release via action on the gonadotropin-releasing hormone (GnRH) system and the gonadotrope in various vertebrates. GnIH peptides identified in birds and mammals have a common LPXRF-amide (X = L or Q) motif at the C-terminus and inhibit pituitary gonadotropin secretion. However, the function and structure of GnIH peptides are diverse in fish. Goldfish GnIHs possessing a C-terminal LPXRF-amide motif have both stimulatory and inhibitory effects on gonadotropin synthesis or release. The C-terminal sequence of grass puffer and medaka GnIHs are MPQRF-amide. To investigate the evolutionary origin of GnIH and its ancestral structure and function, we searched for GnIH in agnathans, the most ancient lineage of vertebrates. We identified GnIH precursor gene and mature GnIH peptides with C-terminal QPQRF-amide or RPQRF-amide from the brain of sea lamprey. Lamprey GnIH fibers were in close proximity to GnRH-III neurons. Further, one of lamprey GnIHs stimulated the expression of lamprey GnRH-III peptide in the hypothalamus and gonadotropic hormone β mRNA expression in the pituitary. We further identified the ancestral form of GnIH, which had a C-terminal RPQRF-amide, and its receptors in amphioxus, the most basal chordate species. The amphioxus GnIH inhibited cAMP signaling in vitro. In sum, the original forms of GnIH may date back to the time of the emergence of early chordates. GnIH peptides may have had various C-terminal structures slightly different from LPXRF-amide in basal chordates, which had stimulatory and/or inhibitory functions on reproduction. The C-terminal LPXRF-amide structure and its inhibitory function on reproduction may be selected in later-evolved vertebrates, such as birds and mammals.
Collapse
Affiliation(s)
- Tomohiro Osugi
- Laboratory of Integrative Brain Sciences, Department of Biology, Center for Medical Life Science, Waseda University Tokyo, Japan
| | - Takayoshi Ubuka
- Laboratory of Integrative Brain Sciences, Department of Biology, Center for Medical Life Science, Waseda University Tokyo, Japan
| | - Kazuyoshi Tsutsui
- Laboratory of Integrative Brain Sciences, Department of Biology, Center for Medical Life Science, Waseda University Tokyo, Japan
| |
Collapse
|
248
|
Bartzen-Sprauer J, Klosen P, Ciofi P, Mikkelsen JD, Simonneaux V. Photoperiodic co-regulation of kisseptin, neurokinin B and dynorphin in the hypothalamus of a seasonal rodent. J Neuroendocrinol 2014; 26:510-20. [PMID: 24935671 DOI: 10.1111/jne.12171] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 12/11/2022]
Abstract
In many species, sexual activity varies on a seasonal basis. Kisspeptin (Kp), a hypothalamic neuropeptide acting as a strong activator of gonadotrophin-releasing hormone neurones, plays a critical role in this adaptive process. Recent studies report that two other neuropeptides, namely neurokinin B (NKB) and dynorphin (DYN), are co-expressed with Kp (and therefore termed KNDy neurones) in the arcuate nucleus and that these peptides are also considered to influence GnRH secretion. The present study aimed to establish whether hypothalamic NKB and DYN expression is photoperiod-dependent in a seasonal rodent, the Syrian hamster, which exhibits robust seasonal rhythms in reproductive activity. The majority of Kp neurones in the arcuate nucleus co-express NKB and DYN and the expression of all three peptides is decreased under a short (compared to long) photoperiod, leading to a 60% decrease in the number of KNDy neurones under photo-inhibitory conditions. In seasonal rodents, RFamide-related peptide (RFRP) neurones of the dorsomedial hypothalamus are also critical for seasonal reproduction. Interestingly, NKB and DYN are also expressed in the dorsomedial hypothalamus but do not co-localise with RFRP-immunoreactive neurones, and the expression of both NKB and DYN is higher under a short photoperiod, which is opposite to the short-day inhibition of RFRP expression. In conclusion, the present study shows that NKB and DYN display different photoperiodic variations in the Syrian hamster hypothalamus. In the arcuate nucleus, NKB and DYN, together with Kp, are down-regulated under a short photoperiod, whereas, in the dorsomedial hypothalamus, NKB and DYN are up-regulated under a short photoperiod.
Collapse
Affiliation(s)
- J Bartzen-Sprauer
- Neurobiologie des Rythmes, Institut des Neurosciences Cellulaires et Intégratives, CNRS 3212, Université de Strasbourg, Strasbourg, France
| | | | | | | | | |
Collapse
|
249
|
León S, García-Galiano D, Ruiz-Pino F, Barroso A, Manfredi-Lozano M, Romero-Ruiz A, Roa J, Vázquez MJ, Gaytan F, Blomenrohr M, van Duin M, Pinilla L, Tena-Sempere M. Physiological roles of gonadotropin-inhibitory hormone signaling in the control of mammalian reproductive axis: studies in the NPFF1 receptor null mouse. Endocrinology 2014; 155:2953-65. [PMID: 24823392 DOI: 10.1210/en.2014-1030] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
RF-amide-related peptide-3 (RFRP-3), the mammalian ortholog of the avian gonadotropin-inhibiting hormone (GnIH), operates via the NPFF1 receptor (NPFF1R) to repress the reproductive axis, therefore acting as counterpart of the excitatory RF-amide peptide, kisspeptin (ligand of Gpr54). In addition, RFRP-3 modulates feeding and might contribute to the integrative control of energy homeostasis and reproduction. Yet, the experimental evidence supporting these putative functions is mostly indirect, and the physiological roles of RFRP-3 remain debatable and obscured by the lack of proper analytical tools and models. To circumvent these limitations, we characterize herein the first mouse line with constitutive inactivation of NPFF1R. Ablation of NPFF1R did not compromise fertility; rather, litters from NPFF1R null mice were larger than those from wild-type animals. Pubertal timing was not altered in NPFF1R deficient mice; yet, pre-pubertal knockout (KO) males displayed elevated LH levels, which normalized after puberty. Adult NPFF1R null male mice showed increased Kiss1 expression in the hypothalamic arcuate nucleus, higher serum FSH levels, and enhanced LH responses to GnRH. However, genetic elimination of NPFF1R was unable to reverse the state of hypogonadism caused by the lack of kisspeptin signaling, as revealed by double NPFF1R/Gpr54 KO mice. NPFF1R null mice displayed altered feedback responses to gonadal hormone withdrawal. In addition, metabolic challenges causing gonadotropin suppression, such as short-term fasting and high-fat diet, were less effective in dampening LH secretion in NPFF1R-deficient male mice, suggesting that absence of this inhibitory pathway partially prevented gonadotropin suppression by metabolic stress. Our data are the first to document the impact of elimination of GnIH signaling on reproductive parameters and their modulation by metabolic challenges. Whereas, in keeping with its inhibitory role, the NPFF1R pathway seems dispensable for preserved puberty and fertility, our results surface different alterations due to the lack of GnIH signaling that prominently include changes in the sensitivity to fasting- and obesity-associated hypogonadotropism.
Collapse
Affiliation(s)
- Silvia León
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, 14004 Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Intraamygdaloid microinjection of RFamide-related peptide-3 decreases food intake in rats. Brain Res Bull 2014; 107:61-8. [DOI: 10.1016/j.brainresbull.2014.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/13/2014] [Accepted: 07/05/2014] [Indexed: 11/23/2022]
|