Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 239(1): 81­91, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30307156

ABSTRACT

RFamide-related peptide (RFRP-3) is a regulator of GnRH secretion from the brain, but it can also act in human ovary to influence steroidogenesis. We aimed to study the putative local role of RFRP-3 in the ovary and its potential participation in the development of a polycystic ovary phenotype induced by chronic sympathetic stress (cold stress). We used adult Sprague­Dawley rats divided into control and stressed groups. In both groups, we studied the effect of intraovarian exposure to RFRP-3 on follicular development and plasma ovarian steroid concentrations. We also tested the effect of RFRP-3 on ovarian steroid production in vitro. Chronic in vivo intraovarian exposure to RFRP-3 decreased basal testosterone concentrations and cold stress-induced progesterone production by the ovary. In vitro, RFRP-3 decreased hCG-induced ovarian progesterone and testosterone secretion. Immunohistochemistry and mRNA expression analysis showed a decrease in Rfrp and expression of its receptor in the ovary of stressed rats, a result which is in line with the increased testosterone levels found in stressed rats. In vivo application of RFRP-3 recovered the low levels of secondary and healthy antral follicles found in stressed rats. Taken together, our data indicate a previously unknown response of hypothalamic and ovarian RFRP-3 to chronic cold stress, influencing ovarian steroidogenesis and follicular dynamics. Thus, it is likely that RFRP-3 modulation in the ovary is a key component of development of the polycystic ovary phenotype.


Subject(s)
Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Ovary/metabolism , Polycystic Ovary Syndrome/metabolism , Animals , Cold Temperature , Female , Polycystic Ovary Syndrome/etiology , Progesterone/blood , Rats, Sprague-Dawley , Receptors, Neuropeptide/metabolism , Stress, Physiological , Testosterone/blood
2.
J Neuroendocrinol ; 30(7): e12597, 2018 07.
Article in English | MEDLINE | ID: mdl-29624758

ABSTRACT

At the turn of the millennium, a neuropeptide with pronounced inhibitory actions on avian pituitary gonadotrophin secretion was identified and named gonadotrophin-inhibitory hormone (GnIH). Across bird species, GnIH acts at the level of the pituitary and the gonadotrophin-releasing hormone (GnRH) neuronal system to inhibit reproduction. Subsequent to this initial discovery, orthologues of GnIH have been identified and characterised across a broad range of species. In many vertebrates, the actions of GnIH and its orthologues serve functional roles analogous to those seen in birds. In other cases, GnIH and its orthologues exhibit more diverse actions dependent on sex, species, season and reproductive condition. The present review highlights the discovery and functional implications of GnIH across species, focusing on research domains in which the significance of this neuropeptide has been explored most.


Subject(s)
Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Reproduction/physiology , Stress, Psychological/metabolism , Animals , Birds , Mammals
3.
Gen Comp Endocrinol ; 237: 140-146, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27591072

ABSTRACT

Gonadotropin inhibitory hormone (GnIH) exerts powerful inhibitory effects on various levels of the vertebrate hypothalamic-pituitary-gonadal (reproductive) axis, yet little is known of how it might change naturally over the course of reproduction. We characterized patterns of hypothalamic GnIH cell abundance over the reproductive period in two popular models used for the study of reproductive endocrinology: European starlings (Sturnus vulgaris) and Sprague-Dawley rats (Rattus norvegicus). We also examined the effects on an unpredictable change in the environment on GnIH cell abundance during the reproductive period, specifically during the period of parental care, by simulating a nest predation event and removing eggs/pups. In both species, we report changes in GnIH cell abundance are occurring at similar reproductive time points but are not always directionally parallel; this may be due to a difference in life histories and physiology mediating parental care. We discovered that cells immunoreactive for the GnIH peptide in male and female starlings are most highly abundant on the first day of incubation and the first day after the first chick hatches. Conversely in rats, GnIH cell abundance decreases in dams on the first day after pups are born. In both male and female starlings and female rats, GnIH cell abundance increases in response to egg/pup loss, indicating that GnIH responds to an unpredictable change in the environment in a potentially conserved fashion. These changes in GnIH cell abundance during the reproductive period inspire further investigation of its adaptive role in reproductive physiological events and behaviors, especially parental care.


Subject(s)
Gonadotropins/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Reproduction/physiology , Starlings/metabolism , Animals , Female , Hypothalamus/cytology , Male , Peptides/metabolism , Pituitary Gland/metabolism , Rats, Sprague-Dawley
4.
J Neuroendocrinol ; 28(7)2016 07.
Article in English | MEDLINE | ID: mdl-27329133

ABSTRACT

Reproduction involves the integration of hormonal signals acting across multiple systems to generate a synchronised physiological output. A critical component of reproduction is the luteinising hormone (LH) surge, which is mediated by oestradiol (E2 ) and neuroprogesterone interacting to stimulate kisspeptin release in the rostral periventricular nucleus of the third ventricle in rats. Recent evidence indicates the involvement of both classical and membrane E2 and progesterone signalling in this pathway. A metabolite of gonadotrophin-releasing hormone (GnRH), GnRH-(1-5), has been shown to stimulate GnRH expression and secretion, and has a role in the regulation of lordosis. Additionally, gonadotrophin release-inhibitory hormone (GnIH) projects to and influences the activity of GnRH neurones in birds. Stress-induced changes in GnIH have been shown to alter breeding behaviour in birds, demonstrating another mechanism for the molecular control of reproduction. Peripherally, paracrine and autocrine actions within the gonad have been suggested as therapeutic targets for infertility in both males and females. Dysfunction of testicular prostaglandin synthesis is a possible cause of idiopathic male infertility. Indeed, local production of melatonin and corticotrophin-releasing hormone could influence spermatogenesis via immune pathways in the gonad. In females, vascular endothelial growth factor A has been implicated in an angiogenic process that mediates development of the corpus luteum and thus fertility via the Notch signalling pathway. Age-induced decreases in fertility involve ovarian kisspeptin and its regulation of ovarian sympathetic innervation. Finally, morphological changes in the arcuate nucleus of the hypothalamus influence female sexual receptivity in rats. The processes mediating these morphological changes have been shown to involve the rapid effects of E2 controlling synaptogenesis in this hypothalamic nucleus. In summary, this review highlights new research in these areas, focusing on recent findings concerning the molecular mechanisms involved in the central and peripheral hormonal control of reproduction.


Subject(s)
Hormones/physiology , Reproduction/physiology , Animals , Humans , Signal Transduction
5.
J Neuroendocrinol ; 27(10): 772-86, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26258544

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.


Subject(s)
Astrocytes/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Hypothalamic Hormones/physiology , Neurons/metabolism , Animals , Female , Male , Rats, Long-Evans
6.
Horm Behav ; 62(2): 173-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22789464

ABSTRACT

Lipopolysaccharide (LPS) is frequently used experimentally to mimic acute infection. Through activation of the host's immune response, an LPS injection has profound effects on the adrenocortical response to stress and on behaviors including reduction in activity, water and food intake, and libido. These behavioral changes occurring during infection are collectively called "sickness behavior." It is thought that adoption of sickness behavior reallocates energy from other fitness-enhancing activities, such as reproduction, for use in the immune response. Although the behavioral effects of LPS treatment are well-known, less information is available regarding the effects of LPS on the brain in terms of controlling reproductive behavior, specifically concerning a newly discovered neuropeptide, gonadotropin-inhibitory hormone (GnIH). This study investigated the effects of an LPS injection on the behavior and the hypothalamic neuropeptides controlling reproduction [GnIH and gonadotropin-releasing hormone (GnRH)] of zebra finches (Taeniopygia guttata). Overall, there was a decrease in activity in birds injected with LPS. The number of GnRH-immunoreactive neurons was significantly reduced in birds injected with LPS when compared to controls, while the number of GnIH-releasing neurons remained unchanged. At the level of gene expression, a similar pattern was found: there was reduced expression of GnRH mRNA in LPS-injected animals, whereas GnIH expression remained unchanged. Plasma testosterone did not change significantly in LPS-injected animals, nor did plasma corticosterone. Taken together, these results indicate a rapid (within 3h) inhibition of the reproductive axis during an immune challenge mimicking an infection, specifically acting on the GnRH system. The present study expands our knowledge on the interaction between the immune system and the reproductive system.


Subject(s)
Avian Proteins , Finches , Gonadotropin-Releasing Hormone , Hypothalamic Hormones , Hypothalamus/drug effects , Lipopolysaccharides/pharmacology , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Finches/genetics , Finches/metabolism , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamic Hormones/genetics , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Injections , Lipopolysaccharides/administration & dosage , Male , Peptide Fragments/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Time Factors
7.
Horm Metab Res ; 44(8): 587-91, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22438212

ABSTRACT

Reproduction is an event that requires the coordination of peripheral organs with the nervous system to ensure that the internal and external environments are optimal for successful procreation of the species. This is accomplished by the hypothalamic-pituitary-gonadal axis that coordinates reproductive behavior with ovulation. The primary signal from the central nervous system is gonadotropin-releasing hormone (GnRH), which modulates the activity of anterior pituitary gonadotropes regulating follicle stimulating hormone (FSH) and luteinizing hormone (LH) release. As ovarian follicles develop they release estradiol, which negatively regulates further release of GnRH and FSH. As estradiol concentrations peak they trigger the surge release of GnRH, which leads to LH release inducing ovulation. Release of GnRH within the central nervous system helps modulate reproductive behaviors providing a node at which control of reproduction is regulated. To address these issues, this review focuses on several critical questions. How is the HPG axis regulated in species with different reproductive strategies? What internal and external conditions modulate the synthesis and release of GnRH? How does GnRH modulate reproductive behavior within the hypothalamus? How does disease shift the activity of the HPG axis?


Subject(s)
Hormones/pharmacology , Reproduction/drug effects , Animals , Behavior/drug effects , Female , Humans , Luteinizing Hormone/metabolism , Ovarian Diseases/physiopathology , Ovulation/drug effects
8.
Genes Brain Behav ; 10(5): 557-64, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21466656

ABSTRACT

Discoveries of how social behavior can influence the plasticity of gonadotropin-releasing hormone (GnRH) have revolutionized the field of behavioral neuroendocrinology by providing new insights into the neural mechanisms controlling behavior. In 2000, the neuropeptide gonadotropin inhibitory hormone (GnIH) was discovered and is changing the way we view how the brain mediates reproduction and associated behaviors. GnIH acts as a reproductive 'pause button', momentarily inhibiting the activity of the reproductive system. However, how GnIH fluctuates naturally in response to social environment is unknown. We examine how the outcome of competition for limited resources needed for reproduction is associated with GnIH. We experimentally manipulated nesting opportunities for pairs of European starlings (Sturnus vulgaris) and examined brain GnIH mRNA and peptide content, as well as GnRH content and plasma testosterone and corticosterone. By limiting the number of nest boxes per enclosure and thus the number of social pairing and nesting opportunities, we observed that birds which outcompeted others for nest boxes ('winners') had significantly fewer numbers of GnIH peptide-producing cells than those without nest boxes ('losers') and this relationship changed with breeding stage. GnRH content, testosterone and corticosterone did not vary with nest box ownership. Thus, while birds appeared reproductively capable across treatments, our data indicate that GnIH may serve as a modulator of reproductive behaviors in response to social environment. Additionally, we provide some evidence of the adaptive value of this mechanism.


Subject(s)
Avian Proteins/metabolism , Hypothalamic Hormones/metabolism , Reproduction/physiology , Sexual Behavior, Animal/physiology , Social Dominance , Social Environment , Animals , Avian Proteins/genetics , Corticosterone/blood , Female , Gonadotropin-Releasing Hormone/blood , Hypothalamic Hormones/genetics , Hypothalamus/metabolism , Male , Starlings , Testosterone/blood
9.
J Neuroendocrinol ; 22(7): 692-700, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20646173

ABSTRACT

To maximise reproductive success, organisms restrict breeding to optimal times of the day or year, when internal physiology and external environmental conditions are suitable for the survival of both parent and offspring. To appropriately coordinate reproductive activity, internal and external standing is communicated to the hypothalamic-pituitary-gonadal axis via a coordinated balance of stimulatory and inhibitory neurochemical systems. The cumulative balance of these mediators ultimately drives the pattern of gonadotrophin-releasing hormone secretion, a neurohormone that stimulates pituitary gonadotrophin secretion. Until 2000, a complementary inhibitor of pituitary gonadotrophin secretion had not been identified. At this time, a novel, avian hypothalamic peptide capable of inhibiting gonadotrophin secretion in cultured quail pituitary cells was uncovered and named gonadotrophin-inhibitory hormone (GnIH). Subsequently, the presence and functional role for the mammalian orthologue of GnIH, RFamide-related peptide, (RFRP-3), was examined, confirming a conserved role for this peptide across several rodent species. To date, a similar distribution and functional role for RFRP-3 have been observed across all mammals investigated, including humans. This overview summarises the role that RFRP-3 plays in mammals and considers the implications and opportunities for further study with respect to reproductive physiology and the neural control of sexual behaviour and motivation.


Subject(s)
Behavior, Animal/physiology , Behavior/physiology , Neuropeptides/metabolism , Reproduction/physiology , Animals , Gonads/metabolism , Humans , Hypothalamus/metabolism , Luteinizing Hormone/metabolism , Pituitary Gland/metabolism , Seasons , Sexual Behavior , Sexual Behavior, Animal
10.
J Neuroendocrinol ; 22(7): 716-27, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20456604

ABSTRACT

Gonadotrophin-releasing hormone (GnRH) is the primary hypothalamic factor responsible for the control of gonadotrophin secretion in vertebrates. However, within the last decade, two other hypothalamic neuropeptides have been found to play key roles in the control of reproductive functions: gonadotrophin-inhibitory hormone (GnIH) and kisspeptin. In 2000, we discovered GnIH in the quail hypothalamus. GnIH inhibits gonadotrophin synthesis and release in birds through actions on GnRH neurones and gonadotrophs, mediated via GPR147. Subsequently, GnIH orthologues were identified in other vertebrate species from fish to humans. As in birds, mammalian and fish GnIH orthologues inhibit gonadotrophin release, indicating a conserved role for this neuropeptide in the control of the hypothalamic-pituitary-gonadal axis across species. Subsequent to the discovery of GnIH, kisspeptin, encoded by the KiSS-1 gene, was discovered in mammals. By contrast to GnIH, kisspeptin has a direct stimulatory effect on GnRH neurones via GPR54. GPR54 is also expressed in pituitary cells, but whether gonadotrophs are targets for kisspeptin remains unresolved. The KiSS-1 gene is also highly conserved and has been identified in mammals, amphibians and fish. We have recently found a second isoform of KiSS-1, designated KiSS-2, in several vertebrates, but not birds, rodents or primates. In this review, we highlight the discovery, mechanisms of action, and functional significance of these two chief regulators of the reproductive axis.


Subject(s)
Glycoproteins/metabolism , Gonadotropins/antagonists & inhibitors , Neuropeptides/metabolism , Reproduction/physiology , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Biological Evolution , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins/genetics , Gonadotropins/metabolism , Humans , Molecular Sequence Data , Neurons/metabolism , Neuropeptides/classification , Neuropeptides/genetics , Phylogeny , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
11.
J Neuroendocrinol ; 21(4): 276-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19210295

ABSTRACT

Gonadotrophin-inhibitory hormone (GnIH) was discovered 8 years ago in birds. Its identification raised the possibility that gonadotrophin-releasing hormone (GnRH) is not the sole hypothalamic neuropeptide that directly influences pituitary gonadotrophin release. Initial studies on GnIH focused on the avian anterior pituitary as comprising the only physiological target of GnIH. There are now several lines of evidence indicating that GnIH directly inhibits pituitary gonadotrophin synthesis and release in birds and mammals. Histological studies on projections from hypothalamic GnIH neurones subsequently implied direct actions of GnIH within the brain and in the periphery. In addition to actions on the pars distalis via the median eminence, GnIH axons and terminals are present in multiple brain areas in birds, and the GnIH receptor is expressed on GnRH-I and -II neurones. Furthermore, we have demonstrated the presence of GnIH and its receptor in avian and mammalian gonads. Thus, GnIH can act directly at multiple levels: within the brain, on the pituitary and in the gonads. In sum, our data indicate that GnIH and its related peptides are important modulators of reproductive function at the level of the GnRH neurone, the gonadotroph and the gonads. Here, we provide an overview of the known levels of GnIH action in birds and mammals. In addition, environmental and physiological factors that are involved in GnIH regulation are reviewed.


Subject(s)
Brain/metabolism , Gonadotropins/metabolism , Gonads/growth & development , Hypothalamic Hormones/metabolism , Pituitary Gland/metabolism , Animals , Brain/growth & development , Humans , Hypothalamus/metabolism , Neurons/metabolism , Periodicity , Reproduction/physiology
12.
J Neuroendocrinol ; 21(4): 271-5, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19207818

ABSTRACT

In vertebrates, the neuropeptide control of gonadotrophin secretion is primarily through the stimulatory action of the hypothalamic decapeptide, gonadotrophin-releasing hormone (GnRH). Gonadal sex steroids and inhibin inhibit gonadotrophin secretion via feedback from the gonads, but a hypothalamic neuropeptide inhibiting gonadotrophin secretion was, until recently, unknown in vertebrates. In 2000, we discovered a novel hypothalamic dodecapeptide that directly inhibits gonadotrophin release in quail and termed it gonadotrophin-inhibitory hormone (GnIH). GnIH acts on the pituitary and GnRH neurones in the hypothalamus via a novel G-protein-coupled receptor for GnIH to inhibit gonadal development and maintenance by decreasing gonadotrophin release and synthesis. The pineal hormone melatonin is a key factor controlling GnIH neural function. GnIH occurs in the hypothalamus of several avian species and is considered to be a new key neurohormone inhibiting avian reproduction. Thus, the discovery of GnIH provides novel directions to investigate neuropeptide regulation of reproduction. This review summarises the discovery, progress and prospects of GnIH, a new key neurohormone controlling reproduction.


Subject(s)
Avian Proteins/isolation & purification , Avian Proteins/metabolism , Gonadotropins/metabolism , Hypothalamic Hormones/isolation & purification , Hypothalamic Hormones/metabolism , Animals , Birds , Coturnix , Gonads/growth & development , Hypothalamus/metabolism , Melatonin/metabolism , Neurons/metabolism , Pituitary Gland/metabolism , Receptors, G-Protein-Coupled/metabolism , Reproduction/physiology
13.
Horm Behav ; 50(5): 762-71, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16919277

ABSTRACT

Many vertebrate species exhibit alternative phenotypes (or morphs), in which one sex displays phenotypic variation equal to or greater than the variation between the sexes. Males in such species typically display differences in reproductive strategies and morphology. Steroid hormones such as testosterone are known modulators of reproductive behavior and morphology and therefore are obvious candidates for the mediation of phenotypic differences between morphs. We conducted a year-round study in the white-throated sparrow (Zonotrichia albicollis) that exhibits alternative phenotypes in plumage coloration and behavior in both sexes: during the breeding season, white-striped males and females are more aggressive and have higher song rates than tan-striped individuals. At the beginning of the breeding season, free-living white-striped males had higher plasma testosterone concentrations than tan-striped males. However, this finding might have been due to different social experiences because captive male morphs sampled at similar times of year did not differ in testosterone concentrations. Captive white-striped males had larger testis and cloacal protuberance sizes than tan-striped males, which might be related to the divergent mating strategies of the morphs. Male morphs showed similar increases in luteinizing hormone following injections of gonadotropin-releasing hormone, but white-striped males showed larger increases in testosterone, indicating differences between morphs in gonadal testosterone production. Females had low concentrations of testosterone, and morphs did not differ. Plasma dehydroepiandrosterone (DHEA) concentrations were elevated in both sexes and morphs during the breeding and non-breeding seasons. These data do not support the hypothesis that testosterone activates behavioral differences between alternative phenotypes in the white-throated sparrow. Alternative testable hypotheses include hormonal effects during early development and direct genetic effects.


Subject(s)
Dehydroepiandrosterone/blood , Estradiol/blood , Phenotype , Pigmentation/physiology , Sexual Behavior, Animal/physiology , Sparrows/physiology , Testosterone/blood , Animals , Body Weight , Cloaca/anatomy & histology , Female , Gonadotropin-Releasing Hormone/pharmacology , Luteinizing Hormone/blood , Male , Sex Characteristics , Sexual Behavior, Animal/drug effects , Testis/anatomy & histology
14.
J Endocrinol ; 182(1): 33-42, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15225129

ABSTRACT

The neuropeptide control of gonadotropin secretion is primarily through the stimulatory action of the hypothalamic decapeptide, GnRH. We recently identified a novel hypothalamic dodecapeptide with a C-terminal LeuPro-Leu-Arg-Phe-NH2 sequence in the domestic bird, Japanese quail (Coturnix japonica). This novel peptide inhibited gonadotropin release in vitro from the quail anterior pituitary; thus it was named gonadotropin-inhibitory hormone (GnIH). GnIH may be an important factor regulating reproductive activity not only in domesticated birds but also in wild, seasonally breeding birds. Thus, we tested synthetic quail GnIH in seasonally breeding wild bird species. In an in vivo experiment, chicken gonadotropin-releasing hormone-I (cGnRH-I) alone or a cGnRH-I/quail GnIH cocktail was injected i.v. into non-breeding song sparrows (Melospiza melodia). Quail GnIH rapidly (within 2 min) attenuated the GnRH-induced rise in plasma LH. Furthermore, we tested the effects of quail GnIH in castrated, photostimulated Gambel's white-crowned sparrows (Zonotrichia leucophrys gambelii), using quail GnIH or saline for injection. Again, quail GnIH rapidly reduced plasma LH (within 3 min) compared with controls. To characterize fully the action of GnIH in wild birds, the identification of their endogenous GnIH is essential. Therefore, in the present study a cDNA encoding GnIH in the brain of Gambel's white-crowned sparrow was cloned by a combination of 3' and 5' rapid amplification of cDNA ends and compared with the quail GnIH cDNA previously identified. The deduced sparrow GnIH precursor consisted of 173 amino acid residues, encoding one sparrow GnIH and two sparrow GnIH-related peptides (sparrow GnIH-RP-1 and GnIH-RP-2) that included Leu-Pro-Xaa-Arg-Phe-NH2 (Xaa=Leu or Gln) at their C-termini. All these peptide sequences were flanked by a glycine C-terminal amidation signal and a single basic amino acid on each end as an endoproteolytic site. Although the homology of sparrow and quail GnIH precursors was approximately 66%, the C-terminal structures of GnIH, GnIH-RP-1 and GnIH-RP-2 were all identical in two species. In situ hybridization revealed the cellular localization of sparrow GnIH mRNA in the paraventricular nucleus (PVN) of the hypothalamus. Immunohistochemical analysis also showed that sparrow GnIH-like immunoreactive cell bodies and terminals were localized in the PVN and median eminence respectively. Thus, only the sparrow PVN expresses GnIH, which appears to be a hypothalamic inhibitory factor for LH release, as evident from our field injections of GnIH into free-living breeding white-crowned sparrows. Sparrow GnIH rapidly (within 2 min) reduced plasma LH when injected into free-living Gambel's white-crowned sparrows on their breeding grounds in northern Alaska. Taken together, our results indicate that, despite amino acid sequence differences, quail GnIH and sparrow GnIH have similar inhibitory effects on the reproductive axis in wild sparrow species. Thus, GnIH appears to be a modulator of gonadotropin release.


Subject(s)
Birds/metabolism , Brain/metabolism , Peptide Hormones/genetics , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Base Sequence , Chickens , Coturnix , DNA, Complementary/analysis , Immunohistochemistry/methods , In Situ Hybridization/methods , Luteinizing Hormone/blood , Male , Molecular Sequence Data , Orchiectomy , Peptide Hormones/metabolism , Quail , Recombinant Proteins/pharmacology , Sequence Alignment
15.
J Neuroendocrinol ; 15(8): 794-802, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12834441

ABSTRACT

Gonadotropin-releasing hormone (GnRH) regulates reproduction in all vertebrates. Until recently, an antagonistic neuropeptide for gonadotropin was unknown. The discovery of an RFamide peptide in quail that inhibits gonadotropin release in vitro raised the possibility of direct hypothalamic inhibition of gonadotropin release. This peptide has now been named gonadotropin-inhibitory hormone (GnIH). We investigated GnIH presence in the hypothalamus of two seasonally breeding songbird species, house sparrows (Passer domesticus) and song sparrows (Melospiza melodia). Using immunocytochemistry (ICC), GnIH-containing neurones were localized in both species in the paraventricular nucleus, with GnIH-containing fibres visible in multiple brain locations, including the median eminence and brainstem. Double-label ICC with light microscopy and fluorescent ICC with confocal microscopy indicate a high probability of colocalization of GnIH with GnRH neurones and fibres within the avian brain. It is plausible that GnIH could be acting at the level of the hypothalamus to regulate gonadotropin release as well as at the pituitary gland. In a photoperiod manipulation experiment, GnIH-containing neurones were larger in birds at the termination of the breeding season than at other times, consistent with a role for this neuropeptide in the regulation of seasonal breeding. We have yet to elucidate the dynamics of GnIH synthesis and release at different times of year, but the data imply temporal regulation of this peptide. In summary, GnIH has the potential to regulate gonadotropin release at more than one level, and its distribution is suggestive of multiple regulatory functions in the central nervous system.


Subject(s)
Avian Proteins , Gonadotropin-Releasing Hormone/analysis , Hypothalamic Hormones/analysis , Median Eminence/chemistry , Reproduction/physiology , Animals , Estrus/physiology , Female , Immunohistochemistry , Male , Median Eminence/cytology , Microscopy, Confocal , Microscopy, Fluorescence , Neurons/chemistry , Photoperiod , Songbirds
16.
J Neuroendocrinol ; 14(10): 805-13, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12372005

ABSTRACT

In songbirds, the initiation of song behaviour and the neural substrate of this system are highly influenced by gonadal steroids. Receptors for gonadal steroid hormones, such as androgens and oestrogens, have been localized within select nuclei of the song system. An important step in steroid receptor action is the recruitment of nuclear receptor coactivators. The coactivator, cAMP response element binding protein (CREB)-binding protein (CBP), has been implicated in both androgen and oestrogen receptor transactivation. Although the role of CBP in transcriptional mechanisms has been widely studied, little is known about CBP expression in the brain. The association between the distribution of CBP and oestrogen receptors in the hippocampus has been related to long-term memory. However, the distribution of brain CBP has not been related to the expression of gonadal steroid receptors in a system as relevant to reproductive behaviour as the avian song system. Western immunoblotting of European starling (Sturnus vulgaris) brain tissue reveals a band at 265 kDa. Immunohistochemical localization of CBP in starling brain indicates wide, but heterogeneous expression. CBP-immunoreactive (CBP-ir) cells define the boundaries of song control nuclei. In HVc (sometimes called the High Vocal Center) and the robust nucleus of the archistriatum (RA), there is a higher density of CBP-ir cells within the boundaries of these nuclei than in adjacent neostriatum or archistriatum, for HVc and RA, respectively. We also report that the distribution of CBP-ir cells varies among different nuclei within the song control system. CBP-ir cells within area X (also a part of the song system) and HVc are densely packed into clusters, whereas cells can be easily discriminated in RA. CBP is also highly expressed in hypothalamic areas, indicating that areas rich in steroid receptors also contain CBP. These data suggest that CBP is important for modulating transcriptional activities in the song system and other sites in the songbird brain that express gonadal steroid receptors.


Subject(s)
Hypothalamus/chemistry , Hypothalamus/physiology , Nuclear Proteins/analysis , Songbirds/physiology , Trans-Activators/analysis , Vocalization, Animal/physiology , Age Factors , Animals , Blotting, Western , CREB-Binding Protein , Immunohistochemistry , Male , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/physiology , Preoptic Area/chemistry , Preoptic Area/physiology , Ventromedial Hypothalamic Nucleus/chemistry , Ventromedial Hypothalamic Nucleus/physiology
17.
J Biol Rhythms ; 16(4): 365-80, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11506381

ABSTRACT

This review examines how birds use the annual cycle in photoperiod to ensure that seasonal events--breeding, molt, and song production--happen at the appropriate time of year. Differences in breeding strategies between birds and mammals reflect basic differences in biology. Avian breeding seasons tend to be of shorter duration and more asymmetric with respect to changes in photoperiod. Breeding seasons can occur at the same time each year (predictable) or at different times (opportunistic), depending on the food resource. In all cases, there is evidence for involvement of photoperiodic control, nonphotoperiodic control, and endogenous circannual rhythmicity. In predictable breeders (most nontropical species), photoperiod is the predominant proximate factor. Increasing photoperiods of spring stimulate secretion of gonadotropin-releasing hormone (GnRH) and consequent gonadal maturation. However, breeding ends before the return of short photoperiods. This is the consequence of a second effect of long photoperiods--the induction of photorefractoriness. This dual role of long photoperiods is required to impart the asymmetry in breeding seasons. Typically, gonadal regression through photorefractoriness is associated with a massive decrease in hypothalamic GnRH, essentially a reversal to a pre-pubertal condition. Although breeding seasons are primarily determined by photoperiodic control of GnRH neurons, prolactin may be important in determining the exact timing of gonadal regression. In tropical and opportunistic breeders, endogenous circannual rhythmicity may be more important. In such species, the reproductive system remains in a state of "readiness to breed" for a large part of the year, with nonphotic cues acting as proximate cues to time breeding. Circannual rhythmicity may result from a temporal sequence of different physiological states rather than a molecular or cellular mechanism as in circadian rhythmicity. Avian homologues of mammalian clock genes Per2, Per3, Clock, bmal1, and MOP4 have been cloned. At the molecular level, avian circadian clocks appear to function in a similar manner to those of mammals. Photoperiodic time measurement involves interaction between a circadian rhythm of photoinducibility and, unlike mammals, deep brain photoreceptors. The exact location of these remains unclear. Although the eyes and pineal generate a daily cycle in melatonin, this photoperiodic signal is not used to time seasonal breeding. Instead, photoperiodic responses appear to involve direct interaction between photoreceptors and GnRH neurons. Thyroid hormones are required in some way for this system to function. In addition to gonadal function, song production is also affected by photoperiod. Several of the nuclei involved in the song system show seasonal changes in volume, greater in spring than in the fall. The increase in volume is, in part, due to an increase in cell number as a result of neurogenesis. There is no seasonal change in the birth of neurons but rather in their survival. Testosterone and melatonin appear to work antagonistically in regulating volume.


Subject(s)
Birds/physiology , Chronobiology Phenomena/physiology , Photoperiod , Seasons , Animals , Hormones/physiology , Sexual Behavior, Animal/physiology , Vocalization, Animal/physiology
18.
Microsc Res Tech ; 53(1): 63-71, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11279671

ABSTRACT

Birds, unlike mammals, do not use the annual profile of pineal melatonin secretion to coordinate their reproductive efforts with a favorable time of year. Melatonin in birds mediates the entrainment of circadian activity rhythms, and thus helps to time hatching of eggs and facilitate migration. However, the role of melatonin as a reliable indicator of day length for seasonal processes has remained equivocal for many years. Recently, the influence of melatonin on two physiological processes involved in aspects of seasonal reproduction has been identified in European starlings: 1) the regulation of seasonal changes in immune function, and 2) neuroplasticity in the song control system. Melatonin can enhance cell-mediated immune function and acts as an inhibitory hormone on the song control system. Melatonin receptor (MelR) density in a forebrain song control nucleus, Area X, is regulated as a function of reproductive state; there is marked downregulation of MelR in Area X during the breeding season in starlings. Seasonal regulation of immune function and neural plasticity within the song control system, and the efficacy of the action of melatonin on these two processes, appears to be modified by the same central, thyroid-dependent mechanism that controls the reproductive state of birds. These data indicate that the interaction of day length and hormones of different classes affects the ability of melatonin to affect seasonal processes in birds. The downstream consequences of MelR regulation within the song control system are discussed with regard to the cellular action of melatonin and its possible interaction with immediate-early genes and transcription factors.


Subject(s)
Birds/physiology , Melatonin/physiology , Seasons , Animals , Female , Male , Reproduction/physiology , Songbirds/physiology
19.
J Neuroendocrinol ; 12(8): 745-52, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10929086

ABSTRACT

Melatonin was recently identified as playing a role in fine-tuning the effects of gonadal steroids in the regulation of seasonal neuroplasticity within the telencephalic song control system of European starlings. The present study investigated possible seasonal regulation of melatonin receptors (MelR) within the starling song control system, in the presence or absence of gonadal steroids. Brains were sampled from photosensitive starlings exposed to short days, photostimulated starlings exposed to long days and photorefractory starlings also exposed to long days. Each condition contained a group of gonad-intact birds and a group of castrated birds. Melatonin receptor distribution was assessed in vitro by 125-iodomelatonin (IMEL) receptor autoradiography. In general, MelR distribution was similar to that described in other songbird species. However, there was a striking downregulation of MelR in the song control nucleus Area X of intact and castrated photostimulated birds on long days compared to their photorefractory counterparts on the same long days and to the short-day groups. Downregulation of MelR occurred independently of gonadal steroids. Nevertheless, superimposed on this general pattern of MelR downregulation during photostimulation, IMEL binding was observed in a medial subdivision of Area X when gonadal steroids were present. Downregulation of MelR in Area X during the short breeding season has implications for seasonal regulation of the song control system. Subsequent upregulation of MelR as birds become photorefractory, in the absence of any change in photoperiod, gonadal steroids or melatonin signal is the first description of photoperiod-independent regulation of MelR in adults of any vertebrate class.


Subject(s)
Photoperiod , Prosencephalon/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Songbirds/metabolism , Animals , Binding, Competitive , Castration , Iodine Radioisotopes , Male , Melatonin/analogs & derivatives , Melatonin/metabolism , Receptors, Cell Surface/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Melatonin , Testosterone/blood , Vocalization, Animal/physiology
20.
J Exp Zool ; 287(1): 74-9, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10861552

ABSTRACT

Exposure of starlings to long days initially causes reproductive maturation, but eventually leads to photorefractoriness. During photorefractoriness, gonadotrophin-releasing hormone (GnRH) decreases in the GnRH cell bodies and fibers emanating from these to the median eminence, circulating gonadotrophin concentrations decrease to a minimum, and the gonads regress. Thyroidectomy profoundly affects these photoperiodic responses. In chronically thyroidectomized starlings, gonadal responses to changes in day length are attenuated. This investigation was conducted to determine whether, in the absence of gonadal responsiveness, the GnRH system of chronically thyroidectomized starlings responds to changes in day length. Two groups of thyroidectomized male starlings were transferred from short days (8L:16D) to long days (18L:6D) for four weeks, and testicular volume increased. One group was kept on long days (TxLD) and the other was returned to short days (TxSD). Testicular volume did not decrease in the TxSD group. The GnRH neurons of the two thyroidectomized groups were compared to those of two groups of intact starlings, one of them on long days and photorefractory (ILD), the other on short days and photosensitive (ISD). Group ILD had lower numbers of GnRH-stained cells than groups TxLD, TxSD and ISD, which did not differ in this respect. Similar differences were observed for GnRH cell size in the pre-optic area (POA) and for density of staining of GnRH fibers in the median eminence. The results confirm that thyroidectomy attenuates gonadal responses to change in day length and suggest that this results from an effect upon the central nervous system rather than a peripheral effect.


Subject(s)
Birds/physiology , Gonadotropin-Releasing Hormone/metabolism , Median Eminence/metabolism , Neurons/metabolism , Photoperiod , Animals , Cell Count , Fluorescent Antibody Technique, Indirect , Image Processing, Computer-Assisted , Male , Median Eminence/cytology , Neurons/cytology , Organ Size , Testis/anatomy & histology , Thyroid Gland/physiology , Thyroidectomy
SELECTION OF CITATIONS
SEARCH DETAIL
...