Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Neurotox Res ; 40(4): 1029-1042, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35639248

ABSTRACT

Despite the fact that the brain is susceptible to neurotoxicity induced by cadmium (Cd), the effects of Cd on the neuroanatomical development in the hypothalamus and regulatory mechanisms of the hypothalamic-pituitary-gonadal (HPG) axis are not fully understood. To clarify this issue, we investigated the effects of 25 mg/kg BW/day cadmium chloride (CdCl2) on neuroanatomical alterations in the hypothalamus of prepubertal female rats. Twenty-four Sprague-Dawley rats were randomly assigned to two groups (n = 12), and CdCl2 was administered via gavage from postnatal days (PND) 21 to PND35. The results of the stereological analysis demonstrated that prepubertal exposure to Cd reduced the number of neurons and oligodendrocytes in the arcuate (ARC) and dorsomedial hypothalamus nucleus (DMH) nuclei. In contrast, Cd exposure increased the number of microglial cells in the ARC and DMH nuclei. Cd exposure decreased the mRNA levels of gonadotropin-releasing hormone (GnRH) and increased the mRNA levels of RFamide-related peptide (RFRP-3), but not kisspeptin (Kiss1) in the hypothalamus. Moreover, hormonal assay showed that Cd exposure caused a reduction in the concentration of gonadotropins: luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in serum. Immunohistochemical expression of RFRP-3 in neuronal cell bodies demonstrated that the mean number of RFRP-3 expressing neurons in the DMH nucleus of cadmium-treated rats was dramatically higher than the vehicle group. Overall, exposure to Cd during the prepubertal period alters the population of neurons and glial cell types in the hypothalamus. Additionally, Cd exposure disrupts the regulatory mechanisms of the HPG axis.


Subject(s)
Cadmium , Hypothalamus , Neuroglia , Animals , Female , Rats , Cadmium/toxicity , Hypothalamus/metabolism , Rats, Sprague-Dawley , RNA, Messenger/metabolism
2.
Front Neuroendocrinol ; 65: 100979, 2022 04.
Article in English | MEDLINE | ID: mdl-35122778

ABSTRACT

This article is an amalgamation of the current status of RFRP-3 (GnIH) in reproduction and its association with the nutrition and stress-mediated changes in the reproductive activities. GnIH has been demonstrated in the hypothalamus of all the vertebrates studied so far and is a well-known inhibitor of GnRH mediated reproduction. The RFRP-3 neurons interact with the other hypothalamic neurons and the hormonal signals from peripheral organs for coordinating the nutritional, stress, and environmental associated changes to regulate reproduction. RFRP-3 has also been shown to regulate puberty, reproductive cyclicity and senescence depending upon the nutritional status. A favourable nutritional status and the environmental cues which are permissive for the successful breeding and pregnancy outcome keep RFRP-3 level low, whereas unfavourable nutritional status and stressful conditions increase the expression of RFRP-3 which impairs the reproduction. Still our knowledge about RFRP-3 is incomplete regarding its therapeutic application for nutritional or stress-related reproductive disorders.


Subject(s)
Neuropeptides , Nutritional Status , Animals , Female , Hypothalamus/metabolism , Neuropeptides/metabolism , Pregnancy , Reproduction/physiology , Sexual Maturation
3.
Front Neuroendocrinol ; 64: 100954, 2022 01.
Article in English | MEDLINE | ID: mdl-34757092

ABSTRACT

The social environment changes circulating hormone levels and expression of social behavior in animals. Social information is perceived by sensory systems, leading to cellular and molecular changes through neural processes. Peripheral reproductive hormone levels are regulated by activity in the hypothalamic-pituitary-gonadal (HPG) axis. Until the end of the last century, the neurochemical systems that convey social information to the HPG axis were not well understood. Gonadotropin-inhibitory hormone (GnIH) was the first hypothalamic neuropeptide shown to inhibit gonadotropin release, in 2000. GnIH is now regarded as a negative upstream regulator of the HPG axis, and it is becoming increasingly evident that it responds to social cues. In addition to controlling reproductive physiology, GnIH seems to modulate the reproductive behavior of animals. Here, we review studies investigating how GnIH neurons respond to social information and describe the mechanisms through which GnIH regulates social behavior.


Subject(s)
Hypothalamic Hormones , Animals , Gonadotropins/metabolism , Hypothalamic Hormones/metabolism , Hypothalamic Hormones/pharmacology , Hypothalamus/metabolism , Social Interaction , Vertebrates/metabolism
4.
Front Neuroendocrinol ; 64: 100953, 2022 01.
Article in English | MEDLINE | ID: mdl-34757094

ABSTRACT

Under stressful condition, reproductive function is impaired due to the activation of various components of the hypothalamic-pituitaryadrenal (HPA) axis, which can suppress the activity of the hypothalamic-pituitary-gonadal (HPG) axis at multiple levels. A hypothalamic neuropeptide, gonadotropin-inhibitory hormone (GnIH) is a key negative regulator of reproduction that governs the HPG axis. Converging lines of evidence have suggested that different stress types and their duration, such as physical or psychological, and acute or chronic, can modulate the GnIH system. To clarify the sensitivity and reactivity of the GnIH system in response to stress, we summarize and critically review the available studies that investigated the effects of various stressors, such as restraint, nutritional/metabolic and social stress, on GnIH expression and/or its neuronal activity leading to altered HPG action. In this review, we focus on GnIH as the potential novel mediator responsible for stress-induced reproductive dysfunction.


Subject(s)
Hypothalamic Hormones , Neuropeptides , Gonadotropins/metabolism , Hypothalamic Hormones/metabolism , Hypothalamic Hormones/pharmacology , Hypothalamus/metabolism , Neuropeptides/metabolism , Reproduction/physiology
5.
Front Neuroendocrinol ; 63: 100948, 2021 10.
Article in English | MEDLINE | ID: mdl-34678303

ABSTRACT

The hypothalamic-pituitary-gonadal axis is the main system that regulates reproduction in vertebrates through a complex network that involves different neuropeptides, neurotransmitters, and pituitary hormones. Considering that this axis is established early on life, the main goal of the present work is to gather information on its development and the actions of its components during early life stages. This review focuses on fish because their neuroanatomical characteristics make them excellent models to study neuroendocrine systems. The following points are discussed: i) developmental functions of the neuroendocrine components of this network, and ii) developmental disruptions that may impact adult reproduction. The importance of the components of this network and their susceptibility to external/internal signals that can alter their specific early functions and/or even the establishment of the reproductive axis, indicate that more studies are necessary to understand this complex and dynamic network.


Subject(s)
Pituitary Gland , Reproduction , Animals , Fishes , Hypothalamus , Neurosecretory Systems
6.
Mol Biol Rep ; 48(2): 1837-1852, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33566226

ABSTRACT

A hypothalamic neuropeptide, RF-amide related peptide-3 (RFRP-3), the mammalian ortholog of the avian gonadotropin-inhibitory hormone (GnIH) has inhibitory signals for reproductive axis via G-protein coupled receptor 147 in mammals. Moreover, RFRP-3 has orexigenic action but the mechanism involved in energy homeostasis and glucose metabolism is not yet known. Though, the RFRP-3 modulates orexigenic action in co-operation with other neuropeptides, which regulates metabolic cues in the hypothalamus. Administration of GnIH/RFRP-3 suppresses plasma luteinizing hormone, at the same time stimulates feeding behavior in birds and mammals. Likewise, in the metabolically deficient conditions, its expression is up-regulated suggests that RFRP-3 contributes to the integration of energy balance and reproduction. However, in many other metabolic conditions like induced diabetes and high-fat diet obesity, etc. its role is still not clear while, RFRP-3 induces the glucose homeostasis by adipocytes is reported. The physiological role of RFRP-3 in metabolic homeostasis and the metabolic effects of RFRP-3 signaling in pharmacological studies need a detailed discussion. Further studies are required to find out whether RFRP-3 is associated with restricted neuroendocrine function observed in type II diabetes mellitus, aging, or sub-fertility. In this context, the current review is focused on the role of RFRP-3 in the above-mentioned mechanisms. Studies from search engines including PubMed, Google Scholar, and science.gov are included after following set inclusion/exclusion criteria. As a developing field few mechanisms are still inconclusive, however, based on the available information RFRP-3 seems to be a putative tool in future treatment strategies towards metabolic disease.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/drug effects , Gonadotropins/metabolism , Hypothalamus/metabolism , Neuropeptides/metabolism , Reproduction/drug effects , Animals , Diabetes Mellitus, Type 2/drug therapy , Energy Metabolism/genetics , Glucose/metabolism , Homeostasis/drug effects , Humans , Neuropeptides/biosynthesis , Neuropeptides/genetics , Neuropeptides/pharmacology , Receptors, Neuropeptide/metabolism , Reproduction/genetics
7.
Front Neuroendocrinol ; 61: 100900, 2021 04.
Article in English | MEDLINE | ID: mdl-33450199

ABSTRACT

The discovery of novel neurohormones is important for the advancement of neuroendocrinology. In early 1970s, gonadotropin-releasing hormone (GnRH), a hypothalamic neuropeptide that promotes gonadotropin release, was identified to be an endogenous neurohormone in mammals. In 2000, thirty years later, another hypothalamic neuropeptide, gonadotropin-inhibitory hormone (GnIH), that inhibits gonadotropin release, was found in quail. GnIH acts via GPR147 and inhibits gonadotropin release and synthesis and reproductive function in birds through actions on GnRH neurons in the hypothalamus and pituitary gonadotrophs. Later, GnIH was found in other vertebrates including humans. GnIH studies have advanced the progress of reproductive neuroendocrinology. Furthermore, recent GnIH studies have indicated that abnormal changes in GnIH expression may cause pubertal disorder and reproductive dysfunction. Here, we describe GnIH discovery and its impact on the progress of reproductive neuroendocrinology. This review also highlights advancement and perspective of GnIH studies on drug development for pubertal disorder and reproductive dysfunction. (149/150).


Subject(s)
Hypothalamic Hormones , Animals , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins , Humans , Hypothalamus/metabolism , Neurotransmitter Agents
8.
J Exp Zool A Ecol Integr Physiol ; 333(4): 214-229, 2020 04.
Article in English | MEDLINE | ID: mdl-32039555

ABSTRACT

The gonadotropin-releasing hormone-gonadotropin inhibitor (GnRH-GnIH) system in the hypothalamus of mammals is the key factor that controls the entire reproductive system. The aim of this study was to immunolocalize GnIH (RFRP-3) in the hypothalamus during the estrous cycle and to study the effect of putrescine on the expression of GnRH-I and GnIH through both in vivo and in vitro (GT1-7 cells) approach and the circulatory levels of GnRH-I, GnIH, and gonadotropins were also investigated. The study also aims in analyzing all the immunofluorescence images by measuring the relative pixel count of an image. This study showed the effect of putrescine on the morphology of ovary, uterus, and the expression of the steroidogenic acute regulatory protein in the ovary. This study showed GnIH expression was intense during the diestrus and moderate during proestrus and estrus, whereas mild staining during the metestrus. The study further showed that putrescine supplementation to adult female rats increased both GnRH-I expression in the hypothalamus as well as the GnRH-I levels in circulation. The study, for the first time, also showed that putrescine supplementation decreased the expression and release of GnIH. These effects of upregulating GnRH-I expression and downregulating GnIH expression were confirmed by in vitro experiments using GT1-7 cells. Putrescine supplementation also increased the gonadotropin levels in the serum. To summarize, putrescine can regulate the hypothalamic-pituitary-gonadal axis by increasing the GnRH-I, luteinizing hormone, and follicle-stimulating hormone levels and suppressing GnIH levels. This is the first report showing the simultaneous effects of putrescine on the regulation of both GnRH-I and GnIH in the hypothalamus.


Subject(s)
Glycoproteins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/physiology , Putrescine/pharmacology , Animals , Cell Line , Estrous Cycle/drug effects , Estrous Cycle/physiology , Female , Follicle Stimulating Hormone , Gene Expression Regulation/drug effects , Glycoproteins/genetics , Gonadotropin-Releasing Hormone/genetics , Hypothalamic Hormones/genetics , Luteinizing Hormone , Neurons/metabolism , Ovary/drug effects , Protein Transport , Rats , Rats, Wistar , Uterus/drug effects
9.
Gen Comp Endocrinol ; 274: 1-7, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30571962

ABSTRACT

Prolactin-releasing peptide2 (PrRP2) belongs to the RFamide peptide group and is a paralog of prolactin-releasing peptide (PrRP). Recent studies demonstrated that PrRP2, but not PrRP, regulates prolactin release in teleosts. The evolutionary origin of PrRP and PrRP2 dates back to at least early vertebrates because homologs of PrRP/PrRP2 were identified in lampreys, one of the earliest branch of vertebrates class Agnatha. However, PrRP/PrRP2 remains to be identified in hagfish, another representative species of class Agnatha. Here, we examined the distribution of PrRP2 in the brain and pituitary of the inshore hagfish Eptatretus burgeri to obtain further understanding of the neuroendocrine system of PrRP2. PrRP2-immunoreactive (ir) cell bodies were detected in the infundibular nucleus of hypothalamus (HYinf). PrRP2-ir fibers were restricted around PrRP2-ir cell bodies and were not detected in the dorsal wall of the neurohypophysis compared to the abundant PrRP2-ir fiber distribution in the brain and innervation to the pituitary in other vertebrates. To examine possible reciprocal connections of PrRP2 and other neuropeptides, we further conducted dual-label immunohistochemistry of PrRP2 and the PQRFamide (PQRFa) peptide or corticotropin-releasing hormone (CRH). Reciprocal connections are suggested between PrRP2 and PQRFa neurons as well as between PrRP2 and CRH neurons. The present study demonstrates, for the first time, that PrRP2 is expressed in the brain of inshore hagfish. The restricted distribution of PrRP2-ir fibers in the HYinf suggests that PrRP2 does not directly regulate the pituitary gland, but regulates the function of the HYinf where PQRFa and CRH are expressed.


Subject(s)
Brain/metabolism , Hagfishes/metabolism , Immunohistochemistry/methods , Prolactin-Releasing Hormone/metabolism , Animals , Antibody Specificity , Corticotropin-Releasing Hormone/metabolism , Female , Hypothalamus/metabolism , Male , Pituitary Gland/metabolism
10.
Reprod Sci ; 26(9): 1249-1255, 2019 09.
Article in English | MEDLINE | ID: mdl-30458681

ABSTRACT

Kisspeptin, encoded by the Kiss-1 gene, plays a crucial role in reproductive function by governing the hypothalamic-pituitary-gonadal axis. The recently established Kiss-1-expressing cell model mHypoA-50 displays characteristics of neuronal cells of the anteroventral periventricular (AVPV) region of the mouse hypothalamus. Because Kiss-1 gene expression in these cells is upregulated by estradiol (E2), mHypoA-50 cells are regarded as a valuable model for the study of Kiss-1-expressing neurons in the AVPV region. These cells also express RFamide-related peptide-3 (RFRP-3), a mammalian homolog of gonadotropin inhibitory hormone. The RFRP-3 expression in mHypoA-50 cells was increased by melatonin stimulation. In addition, E2 stimulation increased RFRP-3 expression in these cells. Treatment of the mHypoA-50 cells with exogenous RFRP-3 resulted in the increase of Kiss-1 messenger RNA expression within the cells; however, RFRP-3 did not modify gonadotropin-releasing hormone or kisspeptin-induced Kiss-1 gene expression in these cells. In addition, we found that RFRP-3 stimulation increased the expression of corticotropin-releasing hormone, which may be involved in E2-induced positive feedback in mHypoA-50 cells. Our observations suggest that RFRP-3 might be involved in positive feedback regulation by directly or indirectly increasing Kiss-1 gene expression.


Subject(s)
Gene Expression Regulation , Hypothalamus/metabolism , Kisspeptins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Animals , Cell Line , Corticotropin-Releasing Hormone/metabolism , Estradiol/pharmacology , Hypothalamus/drug effects , Kisspeptins/genetics , Melatonin/pharmacology , Mice , Neurons/drug effects , Neuropeptides/genetics , Neuropeptides/pharmacology , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
11.
Gen Comp Endocrinol ; 256: 63-70, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28765073

ABSTRACT

The National Institute for Environmental Studies (NIES) of Japan established a strain of Japanese quail (Coturnix japonica) known as NIES-L by rotation breeding in a closed colony for over 35years; accordingly, the strain has highly inbred-like characteristics. Another strain called NIES-Brn has been maintained by randomized breeding in a closed colony to produce outbred-like characteristics. The current study aimed to characterize intermale aggressive behaviors in both strains and to identify possible factors regulating higher aggression in the hypothalamus, such as sex hormone and neuropeptide expression. Both strains displayed a common set of intermale aggressive behaviors that included pecking, grabbing, mounting, and cloacal contact behavior, although NIES-Brn quail showed significantly more grabbing, mounting, and cloacal contact behavior than did NIES-L quail. We examined sex hormone levels in the blood and diencephalon in both strains. Testosterone concentrations were significantly higher in the blood and diencephalon of NIES-Brn quail compared to NIES-L quail. We next examined gene expression in the hypothalamus of both strains using an Agilent gene expression microarray and real-time RT-PCR and found that gene expression of mesotocin (an oxytocin homologue) was significantly higher in the hypothalamus of NIES-Brn quail compared to NIES-L quail. Immunohistochemistry of the hypothalamus revealed that numbers of large cells (cell area>500µm2) expressing mesotocin were significantly higher in the NIES-Brn strain compared to the NIES-L strain. Taken together, our findings suggest that higher testosterone and mesotocin levels in the hypothalamus may be responsible for higher aggression in the NIES-Brn quail strain.


Subject(s)
Aggression/physiology , Coturnix/physiology , Animals , Coturnix/genetics , Estradiol/blood , Gene Expression Regulation , Hypothalamus/metabolism , Japan , Male , Oxytocin/analogs & derivatives , Oxytocin/genetics , Oxytocin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Species Specificity , Testosterone/blood
12.
Endocrinology ; 159(1): 62-68, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28938445

ABSTRACT

It is known that hypothyroidism delays puberty in mammals. Interaction between the hypothalamo-pituitary-thyroid (HPT) and hypothalamo-pituitary-gonadal (HPG) axes may be important processes in delayed puberty. Gonadotropin-inhibitory hormone (GnIH) is a newly discovered hypothalamic neuropeptide that inhibits gonadotropin synthesis and release in quail. It now appears that GnIH is conserved across various mammals and primates, including humans, and inhibits reproduction. We have further demonstrated that GnIH is involved in pubertal delay induced by thyroid dysfunction in female mice. Hypothyroidism delays pubertal onset with the increase in hypothalamic GnIH expression and the decrease in circulating gonadotropin and estradiol levels. Thyroid status regulates GnIH expression by epigenetic modification of the GnIH promoter region. Furthermore, knockout of GnIH gene abolishes the effect of hypothyroidism on delayed pubertal onset. Accordingly, it is considered that GnIH is a mediator of pubertal disorder induced by thyroid dysfunction. This is a novel function of GnIH that interacts between the HPT-HPG axes in pubertal onset delay. This mini-review summarizes the structure, expression, and function of GnIH and highlights the action of GnIH in pubertal disorder induced by thyroid dysfunction.


Subject(s)
Gonadotropins/antagonists & inhibitors , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Hypothyroidism/physiopathology , Models, Neurological , Neurons/metabolism , Puberty, Delayed/etiology , Animals , Gene Expression Regulation, Developmental , Gonadotropins/metabolism , Humans , Hypothalamus/physiopathology , Hypothyroidism/metabolism , Pituitary Gland/metabolism , Pituitary Gland/physiopathology , Thyroid Gland/metabolism , Thyroid Gland/physiopathology
13.
Sci Rep ; 7(1): 1042, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28432332

ABSTRACT

Thyroid disorders cause abnormal puberty, indicating interactions between the hypothalamus-pituitary-thyroid (HPT) and hypothalamus-pituitary-gonadal (HPG) axes, which are important in pubertal development. The hypothalamic gonadotropin-inhibitory hormone (GnIH) was shown to be decreased in the early prepubertal stage, suggesting the role of GnIH on pubertal onset. Here, we investigated whether thyroid dysfunction affects pubertal onset in female mice via GnIH regulation. Hypothyroidism showed delayed pubertal onset with increased GnIH expression and reduced pituitary-gonadal activity. Remarkably, knockout of GnIH prevented the effect of hypothyroidism to delay the pubertal onset, resulting in indistinguishable pubertal timing in GnIH-knockout female mice between control and hypothyroidism-induced group, indicating that increased GnIH expression induced by hypothyroidism may lead to delayed puberty. In contrast, hyperthyroidism led to a decrease in GnIH expression, however pubertal onset was normal, implying further reduction of the inhibitory GnIH had little effect on the phenotypical change. Critically, thyroid hormone suppressed GnIH expression in hypothalamic explants and GnIH neurons expressed thyroid hormone receptors to convey the thyroid status. Moreover, the thyroid status highly regulated the chromatin modifications of GnIH promoter, H3acetylation and H3K9tri-methylation. These findings indicate a novel function of GnIH to mediate HPT-HPG interactions that contribute to proper pubertal development.


Subject(s)
Gonadotropins/metabolism , Hyperthyroidism/complications , Hypothyroidism/complications , Neuropeptides/metabolism , Puberty, Delayed/etiology , Animals , Female , Gene Knockout Techniques , Hypothalamus/drug effects , Mice , Neuropeptides/genetics , Thyroid Hormones/metabolism
14.
Reprod Toxicol ; 71: 32-41, 2017 08.
Article in English | MEDLINE | ID: mdl-28431984

ABSTRACT

The effect of two thyroid disrupting pesticides (TDPs) mancozeb (MCZ) and imidacloprid (IMI) on the hypothalamic-pituitary-gonadal/testicular (HPG) axis of a seasonally breeding bird, Amandava amandava has been evaluated. Male birds (n=8/group) were exposed to each of the pesticide (0.25% LD50 of respective pesticide) as well as to their two equimixture doses (0.25% of LD50 of each and 0.5% LD50 of each) through food for 30d during pre-breeding stage of the reproductive cycle. Reduction in weight, volume and other histopathological features revealed testicular regression. Suppression of gonadotropin releasing hormone, increased expression of gonadotropin inhibitory hormone in the hypothalamus of exposed groups as well as impairment of plasma levels of the reproduction related hormones indicated the disruption of the HPG axis. The pesticides interference of the thyroid function during the critical phase of reproductive development impaired the HPG axis; more significantly in co-exposed groups suggesting the cumulative toxicity.


Subject(s)
Endocrine Disruptors/toxicity , Maneb/toxicity , Neonicotinoids/toxicity , Nitro Compounds/toxicity , Passeriformes/metabolism , Pesticides/toxicity , Zineb/toxicity , Animals , Estradiol/blood , Hypothalamus/metabolism , Male , Peptide Hormones/blood , Peptide Hormones/metabolism , Pituitary Gland/metabolism , Testis/drug effects , Testis/pathology , Testosterone/blood , Thyroid Gland/metabolism
15.
FASEB J ; 30(6): 2198-210, 2016 06.
Article in English | MEDLINE | ID: mdl-26929433

ABSTRACT

Gonadotropin-inhibitory hormone (GnIH) acts as a negative regulator of reproduction by acting on gonadotropes and gonadotropin-releasing hormone (GnRH) neurons. Despite its functional significance, the molecular mechanism of GnIH action in the target cells has not been fully elucidated. To expand our previous study on GnIH actions in gonadotropes, we investigated the potential signal transduction pathway that conveys the inhibitory action of GnIH in GnRH neurons by using the GnRH neuronal cell line, GT1-7. We examined whether GnIH inhibits the action of kisspeptin and vasoactive intestinal polypeptide (VIP), positive regulators of GnRH neurons. Although GnIH significantly suppressed the stimulatory effect of kisspeptin on GnRH release in hypothalamic culture, GnIH had no inhibitory effect on kisspeptin stimulation of serum response element and nuclear factor of activated T-cell response element activities and ERK phosphorylation, indicating that GnIH may not directly inhibit kisspeptin signaling in GnRH neurons. On the contrary, GnIH effectively eliminated the stimulatory effect of VIP on p38 and ERK phosphorylation, c-Fos mRNA expression, and GnRH release. The use of pharmacological modulators strongly demonstrated the specific inhibitory action of GnIH on the adenylate cyclase/cAMP/protein kinase A pathway, suggesting a common inhibitory mechanism of GnIH action in GnRH neurons and gonadotropes.-Son, Y. L., Ubuka, T., Soga, T., Yamamoto, K., Bentley, G. E., Tsutsui, K. Inhibitory action of gonadotropin-inhibitory hormone on the signaling pathways induced by kisspeptin and vasoactive intestinal polypeptide in GnRH neuronal cell line, GT1-7.


Subject(s)
Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/pharmacology , Neurons/drug effects , Vasoactive Intestinal Peptide/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, fos , Hypothalamus/cytology , Mice , Neurons/physiology , Phosphorylation , Protein Kinase C , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Signal Transduction , Vasoactive Intestinal Peptide/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Gen Comp Endocrinol ; 227: 69-76, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26130239

ABSTRACT

Gonadotropin-inhibitory hormone (GnIH) was discovered in 2000 as a novel hypothalamic neuropeptide that inhibited gonadotropin release in the Japanese quail. GnIH and its orthologs have a common C-terminal LPXRFamide (X=L or Q) motif, and have been identified in vertebrates from agnathans to humans, apart from reptiles. In the present study, we characterized a cDNA encoding GnIH orthologs in the brain of the red-eared slider turtle. The deduced precursor protein consisted of 205 amino-acid residues, encoding three putative peptide sequences that included the LPXRFamide motif at their C-termini. In addition, the precursor sequence was most similar to those of avian species. Immunoaffinity purification combined with mass spectrometry confirmed that three mature peptides were produced in the brain. In situ hybridization and immunohistochemistry showed that turtle GnIH-containing cells were restricted to the periventricular hypothalamic nucleus. Immunoreactive fibers were densely distributed in the median eminence. Thus, GnIH and related peptides may act on the pituitary to regulate pituitary hormone release in turtles as well as other vertebrates.


Subject(s)
Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Protein Precursors/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Turtles/metabolism
17.
J Comp Neurol ; 524(9): 1825-38, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-26518222

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.


Subject(s)
Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Hypothalamus/metabolism , Neuropeptides/metabolism , Receptors, Neuropeptide/metabolism , Sex Characteristics , Analysis of Variance , Animals , Avian Proteins/metabolism , Cricetinae , Female , Hypothalamic Hormones/metabolism , Male , Neurons/metabolism , Neuropeptides/genetics , RNA, Messenger/metabolism , Receptors, Neuropeptide/genetics
18.
Gen Comp Endocrinol ; 227: 27-50, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26409890

ABSTRACT

Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that was isolated from the brains of Japanese quail in 2000, which inhibited luteinizing hormone release from the anterior pituitary gland. Here, we summarize the following fifteen years of researches that investigated on the mechanism of GnIH actions at molecular, cellular, morphological, physiological, and behavioral levels. The unique molecular structure of GnIH peptide is in its LPXRFamide (X=L or Q) motif at its C-terminal. The primary receptor for GnIH is GPR147. The cell signaling pathway triggered by GnIH is initiated by inhibiting adenylate cyclase and decreasing cAMP production in the target cell. GnIH neurons regulate not only gonadotropin synthesis and release in the pituitary, but also regulate various neurons in the brain, such as GnRH1, GnRH2, dopamine, POMC, NPY, orexin, MCH, CRH, oxytocin, and kisspeptin neurons. GnIH and GPR147 are also expressed in gonads and they may regulate steroidogenesis and germ cell maturation in an autocrine/paracrine manner. GnIH regulates reproductive development and activity. In female mammals, GnIH may regulate estrous or menstrual cycle. GnIH is also involved in the regulation of seasonal reproduction, but GnIH may finely tune reproductive activities in the breeding seasons. It is involved in stress responses not only in the brain but also in gonads. GnIH may inhibit male socio-sexual behavior by stimulating the activity of cytochrome P450 aromatase in the brain and stimulates feeding behavior by modulating the activities of hypothalamic and central amygdala neurons.


Subject(s)
Avian Proteins/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Pituitary Gland/metabolism , Reproduction/physiology , Signal Transduction/physiology , Animals , Coturnix/metabolism , Female , Gonads/metabolism , Male , Neurons/metabolism
19.
Neuropeptides ; 48(6): 327-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25305807

ABSTRACT

Gonadotropin-inhibitory hormone (GnIH), a 12 amino acid peptide, is expressed in the avian brain and inhibits luteinizing hormone secretion. Additionally, exogenous injection of GnIH causes increased food intake of chicks although the central mechanism mediating this response is poorly understood. Hence, the purpose of our study was to elucidate the central mechanism of the GnIH orexigenic response using 12 day post hatch layer-type chicks as models. Firstly, via mass spectrometry we deduced the chicken GnIH amino acid sequence: SIRPSAYLPLRFamide. Following this we used chicken GnIH to demonstrate that intracerebroventricular (ICV) injection of 2.6 and 7.8 nmol causes increased food intake up to 150 min following injection with no effect on water intake. The number of c-Fos immunoreactive cells was quantified in appetite-associated hypothalamic nuclei following ICV GnIH and only the lateral hypothalamic area (LHA) had an increase of c-Fos positive neurons. From whole hypothalamus samples following ICV GnIH injection abundance of several appetite-associated mRNA was quantified which demonstrated that mRNA for neuropeptide Y (NPY) was increased while mRNA for proopiomelanocortin (POMC) was decreased. This was not the case for mRNA abundance in isolated LHA where NPY and POMC were not affected but melanin-concentrating hormone (MCH) mRNA was increased. A comprehensive behavior analysis was conducted after ICV GnIH injection which demonstrated a variety of behaviors unrelated to appetite were affected. In sum, these results implicate activation of the LHA in the GnIH orexigenic response and NPY, POMC and MCH are likely also involved.


Subject(s)
Avian Proteins/physiology , Eating , Hypothalamic Hormones/physiology , Hypothalamus/metabolism , Animals , Avian Proteins/chemistry , Avian Proteins/pharmacology , Chickens , Drinking/drug effects , Eating/drug effects , Hypothalamic Hormones/chemistry , Hypothalamic Hormones/pharmacology , Injections, Intraventricular , Male , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism
20.
J Mol Endocrinol ; 52(3): T119-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24532655

ABSTRACT

Neuropeptides possessing the Arg-Phe-NH2 (RFamide) motif at their C-termini (designated as RFamide peptides) have been characterized in a variety of animals. Among these, neuropeptide 26RFa (also termed QRFP) is the latest member of the RFamide peptide family to be discovered in the hypothalamus of vertebrates. The neuropeptide 26RFa/QRFP is a 26-amino acid residue peptide that was originally identified in the frog brain. It has been shown to exert orexigenic activity in mammals and to be a ligand for the previously identified orphan G protein-coupled receptor, GPR103 (QRFPR). The cDNAs encoding 26RFa/QRFP and QRFPR have now been characterized in representative species of mammals, birds, and fish. Functional studies have shown that, in mammals, the 26RFa/QRFP-QRFPR system may regulate various functions, including food intake, energy homeostasis, bone formation, pituitary hormone secretion, steroidogenesis, nociceptive transmission, and blood pressure. Several biological actions have also been reported in birds and fish. This review summarizes the current state of identification, localization, and understanding of the functions of 26RFaQRFP and its cognate receptor, QRFPR, in vertebrates.


Subject(s)
Evolution, Molecular , Neuropeptides/genetics , Receptors, G-Protein-Coupled/genetics , Amino Acid Sequence , Animals , Blood Pressure/genetics , Bone Development/genetics , Eating/genetics , Energy Metabolism/genetics , Humans , Hypothalamus/enzymology , Intracellular Signaling Peptides and Proteins/biosynthesis , Molecular Sequence Data , Neuropeptides/biosynthesis , Nociceptive Pain/genetics , Orexins , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL