Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Neurosci Lett ; 837: 137918, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39096756

ABSTRACT

Neurons co-expressing kisspeptin, neurokinin B, and dynorphin A (KNDy neurons), located in the arcuate nucleus (ARC) of the hypothalamus, are indicated to be the gonadotropin-releasing hormone (GnRH) pulse generator. Dynorphin A is reported to suppress GnRH pulse generator activity. Nalfurafine is a selective agonist of the κ-opioid receptor (KOR), a receptor for dynorphin A, clinically used as an anti-pruritic drug. This study aimed to evaluate the effects of nalfurafine on GnRH pulse generator activity and luteinizing hormone (LH) pulses using female goats. Nalfurafine (0, 2, 4, 8, or 16 µg/head) was intravenously injected into ovariectomized Shiba goats. The multiple unit activity (MUA) in the ARC area was recorded, and plasma LH concentrations were measured 2 and 48 h before and after injection, respectively. The MUA volley interval during 0-2 h after injection was significantly increased in the nalfurafine 8 and 16 µg groups compared with the vehicle group. In 0-2 h after injection, the number of LH pulses was significantly decreased in the nalfurafine 8 and 16 µg groups, and the mean and baseline LH were significantly decreased in all nalfurafine-treated groups (2, 4, 8, and 16 µg) compared with the vehicle group. These results suggest that nalfurafine inhibits the activity of the GnRH pulse generator in the ARC, thus suppressing pulsatile LH secretion. Therefore, nalfurafine could be used as a reproductive inhibitor in mammals.


Subject(s)
Arcuate Nucleus of Hypothalamus , Goats , Gonadotropin-Releasing Hormone , Morphinans , Receptors, Opioid, kappa , Spiro Compounds , Animals , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/metabolism , Female , Spiro Compounds/pharmacology , Spiro Compounds/administration & dosage , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/agonists , Morphinans/pharmacology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Kisspeptins/metabolism , Dynorphins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurokinin B/metabolism
2.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1006562

ABSTRACT

ObjectiveTo explore the comprehensive effects of Qingxin Zishen decoction on the symptom score and neuroendocrine indexes and the mechanism of the decoction in regulating KNDy neurons in the patients with menopausal syndrome. MethodA total of 60 patients with menopausal syndrome due to yin deficiency with effulgent fire who attended the menopausal outpatient of Jiangsu Province Hospital of Chinese Medicine were randomized into an experimental (Qingxin Zishen decoction) group (30 cases) and a control (femoston) group (30 cases). The treatment lasted for 12 weeks in both groups. The two groups were compared in terms of the comprehensive efficacy, frequency and degree of hot flashes and sweating, modified Kupperman score, and the serum levels of hypothalamic peptide kisspeptin, neurokinin B (NKB), dynorphin (Dyn), follicle-stimulating hormone (FSH), and estradiol (E2). Result① Comprehensive efficacy: The comprehensive efficacy of the two groups was comparable. ② Frequency and degrees of hot flashes and sweating: After treatment, the frequency and degrees of hot flashes and sweating in the two groups were reduced (P<0.05) and the control group outperformed the experimental group (P<0.05). ③ Modified Kupperman score and menopausal symptoms: After treatment, the modified Kupperman score decreased in both groups (P<0.05). After 4 weeks of treatment, the experimental group was superior to the control group in terms of the scores of dizziness and headache (P<0.05). ④ Serum levels of sex hormones: After treatment, the serum E2 level elevated and the FSH level lowered in both groups (P<0.05), and the changes were more obvious in the control group (P<0.05). ⑤ Neuroendocrine indexes: After treatment, the serum levels of kisspeptin and NKB in the two groups decreased (P<0.05), and the serum Dyn level in the experimental group increased (P<0.05). Moreover, the experimental group had higher Dyn level than the control group after treatment (P<0.05). ConclusionQingxin Zishen decoction can alleviate hot flashes, sweating, and other symptoms in the women with menopausal syndrome by acting on the KNDy neurons to lower the kisspeptin and NKB levels and elevate the Dyn level. The findings provide new ideas for the clinical treatment of hot flashes in menopause.

3.
Biol Reprod ; 109(5): 654-668, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37665248

ABSTRACT

Kisspeptin (KP, encoded by Kiss1, binding to the Gpr54 receptor) is a neuropeptide conveying information on the metabolic status to the hypothalamic-pituitary-gonadal axis. KP acts together with dynorphin A (encoded by Pdyn) and neurokinin B (encoded by Tac2) to regulate reproduction. KP is crucial for the onset of puberty and is under the control of sirtuin (encoded by Sirt1). We hypothesize that the maternal cafeteria (CAF) diet has adverse effects on the offspring's hormonal, metabolic, and reproductive functions due to sex-specific alterations in the expression of Kiss1, Gpr54, Pdyn, Tac2, and Sirt1 in the hypothalamus, and Kiss1, Gpr54, and Sirt1 in the liver. Rats were fed a CAF diet before pregnancy, during pregnancy, and during lactation. The vaginal opening was monitored. Offspring were sacrificed in three age points: PND 30, PND 35, and PND 60 (females) and PND 40, PND 45, and PND 60 (males). Their metabolic and hormonal status was assessed. mRNA for Kiss1, Gpr54, Pdyn, Tac2, and Sirt1 were measured by real-time PCR in the hypothalamus and/or livers. We found that CAF offspring had lower weight and altered body composition; increased cholesterol and triglyceride levels, sex-specific changes in glucose and insulin levels; sex-dependent changes in Sirt1/Kiss1 mRNA ratio in the hypothalamus; sex-specific alterations in Kiss1 and Sirt1 mRNA in the liver with more diversity in males; and a delayed puberty onset in females. We concluded that the mother's CAF diet leads to sex-specific alterations in metabolic and reproductive outcomes via Kiss1/Gpr54 and Sirt1 systems in offspring.


Subject(s)
Kisspeptins , Sirtuin 1 , Pregnancy , Female , Male , Rats , Animals , Kisspeptins/genetics , Kisspeptins/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sexual Maturation/physiology , Receptors, Kisspeptin-1/genetics , Receptors, Kisspeptin-1/metabolism , Diet , Metabolome , RNA, Messenger/metabolism
5.
Ther Adv Reprod Health ; 17: 26334941231177611, 2023.
Article in English | MEDLINE | ID: mdl-37388717

ABSTRACT

Vasomotor symptoms of menopause (VMS), otherwise known as hot flashes, can significantly impact women's quality of life. Up to 87% of women report hot flashes during or after their menopause transition, and can last for a median duration of 7.4 years. The current mainstay of treatment and the most effective treatment for VMS is hormone therapy with estrogen. However, hormone therapy is not without risk, and the discovery of an effective nonhormonal treatment option with neurokinin B receptor antagonists for VMS provides an encouraging and potentially practice-changing treatment option for all women. This review will discuss the pathophysiology and mechanism of action, as well as review the current compounds in development targeting the neurokinin receptors.

6.
Front Endocrinol (Lausanne) ; 13: 951938, 2022.
Article in English | MEDLINE | ID: mdl-36479214

ABSTRACT

Feedback from oestradiol (E2) plays a critical role in the regulation of major events in the physiological menstrual cycle including the release of gonadotrophins to stimulate follicular growth, and the mid-cycle luteinising hormone (LH) surge that leads to ovulation. E2 predominantly exerts its action via oestrogen receptor-alpha (ERα), however, as gonadotrophin releasing hormone (GnRH) neurons lack ERα, E2-feedback is posited to be indirectly mediated via upstream neurons. Kisspeptin (KP) is a neuropeptide expressed in hypothalamic KP-neurons that control GnRH secretion and plays a key role in the central mechanism regulating the hypothalamic-pituitary-gonadal (HPG) axis. In the rodent arcuate (ARC) nucleus, KP is co-expressed with Neurokinin B and Dynorphin; and thus, these neurons are termed 'Kisspeptin-Neurokinin B-Dynorphin' (KNDy) neurons. ARC KP-neurons function as the 'GnRH pulse generator' to regulate GnRH pulsatility, as well as mediating negative feedback from E2. A second KP neuronal population is present in the rostral periventricular area of the third ventricle (RP3V), which includes anteroventral periventricular (AVPV) nucleus and preoptic area neurons. These RP3V KP-neurons mediate positive feedback to induce the mid-cycle luteinising hormone (LH) surge and subsequent ovulation. Here, we describe the role of KP-neurons in these two regions in mediating this differential feedback from oestrogens. We conclude by considering reproductive diseases for which exploitation of these mechanisms could yield future therapies.


Subject(s)
Kisspeptins , Neurokinin B , Dynorphins , Luteinizing Hormone , Gonadotropin-Releasing Hormone , Neurons
7.
Front Neurosci ; 16: 958377, 2022.
Article in English | MEDLINE | ID: mdl-36033602

ABSTRACT

Endogenous opioid peptides have attracted attention as critical neuropeptides in the central mechanism regulating female reproduction ever since the discovery that arcuate dynorphin neurons that coexpress kisspeptin and neurokinin B (NKB), which are also known as kisspeptin/neurokinin B/dynorphin (KNDy) neurons, play a role as a master regulator of pulsatile gonadotropin-releasing hormone (GnRH) release in mammals. In this study, we first focus on the role of dynorphin released by KNDy neurons in the GnRH pulse generation. Second, we provide a historical overview of studies on endogenous opioid peptides. Third, we discuss how endogenous opioid peptides modulate tonic GnRH/gonadotropin release in female mammals as a mediator of inhibitory internal and external cues, such as ovarian steroids, nutritional status, or stress, on reproduction. Then, we discuss the role of endogenous opioid peptides in GnRH surge generation in female mammals.

8.
Front Endocrinol (Lausanne) ; 13: 925206, 2022.
Article in English | MEDLINE | ID: mdl-35837314

ABSTRACT

The discovery of kisspeptin as a critical central regulatory factor of GnRH release has given people a novel understanding of the neuroendocrine regulation in human reproduction. Kisspeptin activates the signaling pathway by binding to its receptor kisspeptin receptor (KISS1R) to promote GnRH secretion, thereby regulating the hypothalamic-pituitary-gonadal axis (HPG) axis. Recent studies have shown that kisspeptin neurons located in arcuate nucleus (ARC) co-express neurokinin B (NKB) and dynorphin (Dyn). Such neurons are called KNDy neurons. KNDy neurons participate in the positive and negative feedback of estrogen to GnRH secretion. In addition, kisspeptin is a key factor in the initiation of puberty, and also regulates the processes of female follicle development, oocyte maturation, and ovulation through the HPG axis. In male reproduction, kisspeptin also plays an important role, getting involved in the regulation of Leydig cells, spermatogenesis, sperm functions and reproductive behaviors. Mutations in the KISS1 gene or disorders of the kisspeptin/KISS1R system may lead to clinical symptoms such as idiopathic hypogonadotropic hypogonadism (iHH), central precocious puberty (CPP) and female infertility. Understanding the influence of kisspeptin on the reproductive axis and related mechanisms will help the future application of kisspeptin in disease diagnosis and treatment. In this review, we critically appraise the role of kisspeptin in the HPG axis, including its signaling pathways, negative and positive feedback mechanisms, and its control on female and male reproduction.


Subject(s)
Kisspeptins , Semen , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , Kisspeptins/metabolism , Male , Receptors, Kisspeptin-1/genetics , Receptors, Kisspeptin-1/metabolism , Reproduction/physiology
9.
J Neuroendocrinol ; 34(5): e13094, 2022 05.
Article in English | MEDLINE | ID: mdl-35107859

ABSTRACT

The concept that different systems control episodic and surge secretion of gonadotropin-releasing hormone (GnRH) was well established by the time that GnRH was identified and formed the framework for studies of the physiological roles of GnRH, and later kisspeptin. Here, we focus on recent studies identifying the neural mechanisms underlying these two modes of secretion, with an emphasis on their core components. There is now compelling data that kisspeptin neurons in the arcuate nucleus that also contain neurokinin B (NKB) and dynorphin (i.e., KNDy cells) and their projections to GnRH dendrons constitute the GnRH pulse generator in mice and rats. There is also strong evidence for a similar role for KNDy neurons in sheep and goats, and weaker data in monkeys and humans. However, whether KNDy neurons act on GnRH dendrons and/or GnRH soma and dendrites that are found in the mediobasal hypothalamus (MBH) of these species remains unclear. The core components of the GnRH/luteinising hormone surge consist of an endocrine signal that initiates the process and a neural trigger that drives GnRH secretion during the surge. In all spontaneous ovulators, the core endocrine signal is a rise in estradiol secretion from the maturing follicle(s), with the site of estrogen positive feedback being the rostral periventricular kisspeptin neurons in rodents and neurons in the MBH of sheep and primates. There is considerable species variations in the neural trigger, with three major classes. First, in reflex ovulators, this trigger is initiated by coitus and carried to the hypothalamus by neural or vascular pathways. Second, in rodents, there is a time of day signal that originates in the suprachiasmatic nucleus and activates rostral periventricular kisspeptin neurons and GnRH soma and dendrites. Finally, in sheep nitric oxide-producing neurons in the ventromedial nucleus, KNDy neurons and rostral kisspeptin neurons all appear to participate in driving GnRH release during the surge.


Subject(s)
Dendrimers , Gonadotropin-Releasing Hormone , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Dendrimers/metabolism , Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Mice , Neurokinin B/metabolism , Rats , Sheep
10.
Front Neuroendocrinol ; 64: 100968, 2022 01.
Article in English | MEDLINE | ID: mdl-34808231

ABSTRACT

Accumulating findings during the past decades have demonstrated that the hypothalamic arcuate kisspeptin neurons are supposed to be responsible for pulsatile release of gonadotropin-releasing hormone (GnRH) to regulate gametogenesis and steroidogenesis in mammals. The arcuate kisspeptin neurons express neurokinin B (NKB) and dynorphin A (Dyn), thus, the neurons are also referred to as KNDy neurons. In the present article, we mainly focus on the cellular and molecular mechanisms underlying GnRH pulse generation, that is focused on the action of NKB and Dyn and an interaction between KNDy neurons and astrocytes to control GnRH pulse generation. Then, we also discuss the factors that modulate the activity of KNDy neurons and consequent pulsatile GnRH/LH release in mammals.


Subject(s)
Arcuate Nucleus of Hypothalamus , Gonadotropin-Releasing Hormone , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Mammals , Neurokinin B/metabolism , Neurons/metabolism
11.
J Neurosci ; 41(47): 9688-9701, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34654752

ABSTRACT

Kisspeptin (Kiss1) neurons are essential for reproduction, but their role in the control of energy balance and other homeostatic functions remains unclear. High-frequency firing of hypothalamic arcuate Kiss1 (Kiss1ARH) neurons releases kisspeptin into the median eminence, and neurokinin B (NKB) and dynorphin onto neighboring Kiss1ARH neurons to generate a slow EPSP mediated by TRPC5 channels that entrains intermittent, synchronous firing of Kiss1ARH neurons. High-frequency optogenetic stimulation of Kiss1ARH neurons also releases glutamate to excite the anorexigenic proopiomelanocortin (POMC) neurons and inhibit the orexigenic neuropeptide Y/agouti-related peptide (AgRP) neurons via metabotropic glutamate receptors. At the molecular level, the endoplasmic reticulum (ER) calcium-sensing protein stromal interaction molecule 1 (STIM1) is critically involved in the regulation of neuronal Ca2+ signaling and neuronal excitability through its interaction with plasma membrane (PM) calcium (e.g., TRPC) channels. Therefore, we hypothesized that deletion of Stim1 in Kiss1ARH neurons would increase neuronal excitability and their synchronous firing, which ultimately would affect energy homeostasis. Using optogenetics in combination with whole-cell recording and GCaMP6 imaging in slices, we discovered that deletion of Stim1 in Kiss1 neurons significantly increased the amplitude and duration of the slow EPSP and augmented synchronous [Ca2+]i oscillations in Kiss1ARH neurons. Deletion of Stim1 in Kiss1ARH neurons amplified the actions of NKB and protected ovariectomized female mice from developing obesity and glucose intolerance with high-fat dieting (HFD). Therefore, STIM1 appears to play a critical role in regulating synchronous firing of Kiss1ARH neurons, which ultimately affects the coordination between energy homeostasis and reproduction.SIGNIFICANCE STATEMENT Hypothalamic arcuate kisspeptin (Kiss1ARH) neurons are essential for stimulating the pulsatile release of gonadotropin-releasing hormone (GnRH) and maintaining fertility. However, Kiss1ARH neurons appear to be a key player in coordinating energy balance with reproduction. The regulation of calcium channels and hence calcium signaling is critically dependent on the endoplasmic reticulum (ER) calcium-sensing protein stromal interaction molecule 1 (STIM1), which interacts with the plasma membrane (PM) calcium channels. We have conditionally deleted Stim1 in Kiss1ARH neurons and found that it significantly increased the excitability of Kiss1ARH neurons and protected ovariectomized female mice from developing obesity and glucose intolerance with high-fat dieting (HFD).


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism/physiology , Kisspeptins/metabolism , Neurons/metabolism , Obesity/metabolism , Stromal Interaction Molecule 1/metabolism , Animals , Calcium Signaling/physiology , Diet, High-Fat , Excitatory Postsynaptic Potentials/physiology , Female , Green Fluorescent Proteins , Mice
12.
Handb Clin Neurol ; 181: 463-496, 2021.
Article in English | MEDLINE | ID: mdl-34238478

ABSTRACT

Puberty, which in humans is considered to include both gonadarche and adrenarche, is the period of becoming capable of reproducing sexually and is recognized by maturation of the gonads and development of secondary sex characteristics. Gonadarche referring to growth and maturation of the gonads is fundamental to puberty since it encompasses increased gonadal steroid secretion and initiation of gametogenesis resulting from enhanced pituitary gonadotropin secretion, triggered in turn by robust pulsatile GnRH release from the hypothalamus. This chapter reviews the development of GnRH pulsatility from before birth until the onset of puberty. In humans, GnRH pulse generation is restrained during childhood and juvenile development. This prepubertal hiatus in hypothalamic activity is considered to result from a neurobiological brake imposed upon the GnRH pulse generator resident in the infundibular nucleus. Reactivation of the GnRH pulse generator initiates pubertal development. Current understanding of the genetics and physiology of the brake will be discussed, as will hypotheses proposed to account for timing the resurgence in pulsatile GnRH and initiation of puberty. The chapter ends with a discussion of disorders associated with precocious or delayed puberty with a focus on those with etiologies attributed to aberrant GnRH neuron anatomy or function. A pediatric approach to patients with pubertal disorders is provided and contemporary treatments for both precocious and delayed puberty outlined.


Subject(s)
Gonadotropin-Releasing Hormone , Puberty , Child , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamus/metabolism , Neurobiology , Neurons/metabolism
15.
Front Neurosci ; 14: 598707, 2020.
Article in English | MEDLINE | ID: mdl-33343288

ABSTRACT

Neurons co-synthesizing kisspeptin (KP), neurokinin B (NKB), and dynorphin ("KNDy neurons") in the hypothalamic arcuate/infundibular nucleus (INF) form a crucial component of the gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) "pulse generator." The goal of our study was to characterize KP neuron distribution, neuropeptide phenotype and connectivity to GnRH cells in ovariectomized (OVX) dogs and cats with immunohistochemistry on formalin-fixed hypothalamic tissue sections. In both species, KP and NKB neurons occurred in the INF and the two cell populations overlapped substantially. Dynorphin was detected in large subsets of canine KP (56%) and NKB (37%) cells and feline KP (64%) and NKB (57%) cells; triple-labeled ("KNDy") somata formed ∼25% of all immunolabeled neurons. Substance P (SP) was present in 20% of KP and 29% of NKB neurons in OVX cats but not dogs, although 26% of KP and 24% of NKB neurons in a gonadally intact male dog also contained SP signal. Only in cats, cocaine- and amphetamine regulated transcript was also colocalized with KP (23%) and NKB (7%). In contrast with reports from mice, KP neurons did not express galanin in either carnivore. KP neurons innervated virtually all GnRH neurons in both species. Results of this anatomical study on OVX animals reveal species-specific features of canine and feline mediobasal hypothalamic KP neurons. Anatomical and neurochemical similarities to and differences from the homologous KP cells of more extensively studied rodent, domestic and primate species will enhance our understanding of obligate and facultative players in the molecular mechanisms underlying pulsatile GnRH/LH secretion.

16.
BAG, J. basic appl. genet. (Online) ; 31(2): 12-25, Dec. 2020. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1345377

ABSTRACT

ABSTRACT Dynorphin A is an endogenous opioid peptide that is part of the KNDy system in the hypothalamus of mammals. This peptide acts as an inhibitor of the GnRH pulse generation, thus regulating the onset of puberty and reproductive cycles. The PDYN gene encodes the propeptide Prodynorphin, the precursor of Dynorphin A. Despite its physiological relevance, PDYN has not emerged as a candidate gene associated with puberty in genomic association studies conducted in cattle. The present work aimed to search for signatures of selection on the PDYN gene among cattle breeds. To this, the whole genome sequences from 57 samples of ten cattle breeds were used. The samples were grouped based on breed selection history and their productive differences, particularly in terms of sexual precocity. The population structure was analyzed using Principal Component Analyses. To evidence recent selection processes, neutrality tests, such as Tajima's D and Fu & Li's F* and D* were performed in defined functional regions of PDYN. The putative promoter of PDYN showed a population structure that is in agreement with the criteria considered to make the groups. In that region, neutrality tests were consistently negative and resulted in statistically significant for the dairy breeds. Also, these breeds exhibited less variability in the haplotype analyses than the others. The results presented here suggest that regulatory regions of PDYN could be under positive selection, particularly in dairy breeds.


RESUMEN Dinorfina A es un péptido opioide endógeno que forma parte del sistema KNDy en el hipotálamo de mamíferos. Este péptido actúa como inhibidor de la generación de los pulsos de GnRH, regulando así el inicio de la pubertad y los ciclos reproductivos. El gen PDYN codifica el propéptido Prodinorfina, precursor de Dinorfina A. A pesar de su relevancia fisiológica, PDYN no ha surgido como gen candidato asociado a pubertad en estudios de asociación genómicos en bovinos. El presente trabajo tuvo como objetivo buscar huellas de selección en el gen PDYN entre diferentes razas bovinas. Para alcanzarlo se utilizaron secuencias genómicas de 57 muestras de diez razas bovinas. Las muestras fueron agrupadas considerando la historia de selección y las diferencias productivas entre razas, particularmente en términos de precocidad sexual. La estructura poblacional fue analizada usando análisis de componentes principales. Para evidenciar procesos de selección recientes se realizaron pruebas de neutralidad, tales como D de Tajima y F* y D* de Fu & Li, en diferentes regiones funcionales de PDYN. El promotor putativo de PDYN mostró una estructura poblacional que es consistente con los criterios usados para agrupar las razas. En esa región, las pruebas de neutralidad fueron consistentemente negativas y estadísticamente significativas en las razas lecheras. Además, estas razas también exhibieron menor variabilidad en los análisis de haplotipos que las demás razas. Los resultados presentados aquí sugieren que regiones regulatorias de PDYN estarían bajo selección positiva, particularmente en razas bovinas lecheras.

17.
Neuroscience ; 451: 184-196, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33065232

ABSTRACT

KNDy neurons co-expressing kisspeptin (KP), neurokinin B (NKB) and dynorphin A (DYN A) in the arcuate nucleus of the hypothalamus (ARC) are key regulators of reproduction. Their activity is influenced by metabolic and hormonal signals. Previously, we have shown that orchidectomy alters the KP-, NKB-, and DYN A-immunoreactivity in the high-fat diet-induced (HFD) obesity and diabetes type 2 (DM2) models. Considering the potential sex difference in the response of KNDy neurons, we have hypothesized that ovariectomy (OVX) and post-ovariectomy replacement with estradiol (OVX+E2) or estradiol and progesterone (OVX+E2+P4) will also affect these neurons in HFD and DM2 females. Thus, each of these treatment protocols were employed for control, HFD, and DM2 groups of rats leading to nine experimental conditions within which we have determined the number of KP-, NKB-, or DYN-immunoreactive (-ir) neurons and assessed the metabolic and hormonal profiles of the animals. Accordingly: (1) no effects of group and surgery were observed on the number of KP-ir neurons; (2) the overall number of NKB-ir neurons was higher in the OVX+E2+P4 and OVX+E2 animals compared to OVX; (3) overall, the number of DYN A-ir neurons was higher in DM2 vs. control group, and surgery had an effect on the number of DYN A-ir neurons; (4) the metabolic and hormonal profiles were altered in HFD and DM2 animals compared to controls. Current data together with our previously published results indicate sex-specific differences in the response of KNDy neurons to DM2.


Subject(s)
Arcuate Nucleus of Hypothalamus , Diabetes Mellitus, Experimental , Dynorphins , Gonadal Steroid Hormones , Kisspeptins , Neurokinin B , Neurons , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , Female , Gonadotropin-Releasing Hormone/metabolism , Male , Neurokinin B/metabolism , Neurons/metabolism , Obesity , Ovariectomy , Rats
18.
Neurosci Lett ; 736: 135276, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32771877

ABSTRACT

Pulsatile gonadotropin-releasing hormone (GnRH) secretion is essential for regulating reproductive functions in mammals. GnRH pulses are governed by a neural mechanism that is termed the GnRH pulse generator. In the present study, we investigated the role of central calcitonin receptor (CTR) signaling in the regulation of the GnRH pulse generator activity in ovariectomized goats by administering amylin, an endogenous ligand for CTR, into the lateral ventricle. GnRH pulse generator activity was measured using multiple unit activity (MUA) recordings in the mediobasal hypothalamus. We analyzed changes in the interval of characteristic increases in MUA (MUA volleys). The MUA volley interval shortened immediately after amylin administration, followed by prolonged intervals. Double in situ hybridization for KISS1 (kisspeptin gene) and CALCR (CTR gene) revealed that low expression levels of CALCR were found in the arcuate kisspeptin neurons, which is suggested as the main population of neurons, involved in GnRH pulse generator activity. These results suggest that central amylin-CTR signaling has a biphasic role in the regulation of GnRH pulse generator activity by acting on cells other than the arcuate kisspeptin neurons in goats.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Islet Amyloid Polypeptide/administration & dosage , Neurons/drug effects , Animals , Female , Goats , Hypothalamus/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/blood , Neurons/metabolism , Receptors, Calcitonin/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Endocr J ; 67(4): 409-418, 2020 Apr 28.
Article in English | MEDLINE | ID: mdl-31941848

ABSTRACT

Accumulating evidence suggests that kisspeptin neurons in the arcuate nucleus (ARC), which coexpress neurokinin B and dynorphin, are involved in gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) pulse generation, while the anteroventral periventricular nucleus (AVPV) kisspeptin neurons are responsible for GnRH/LH surge generation. The present study aims to examine whether GnRH(1-5), a GnRH metabolite, regulates LH release via kisspeptin neurons. GnRH(1-5) was intracerebroventricularly injected to ovariectomized and estrogen-treated Wistar-Imamichi female rats. Immediately after the central GnRH(1-5) administration at 2 nmol, plasma LH concentration increased, resulting in significantly higher levels of the area under the curve and baseline of plasma LH concentrations compared to vehicle-injected controls. On the other hand, in Kiss1 knockout rats, GnRH(1-5) administration failed to affect LH secretion, suggesting that the facilitatory effect of GnRH(1-5) on LH release is mediated by kisspeptin neurons. Double in situ hybridization (ISH) for Kiss1 and Gpr101, a GnRH(1-5) receptor gene, revealed that few Kiss1-expressing cells coexpress Gpr101 in both ARC and AVPV. On the other hand, double ISH for Gpr101 and Slc17a6, a glutamatergic marker gene, revealed that 29.2% of ARC Gpr101-expressing cells coexpress Slc17a6. Further, most of the AVPV and ARC Kiss1-expressing cells coexpress Grin1, a gene encoding a subunit of NMDA receptor. Taken together, these results suggest that the GnRH(1-5)-GPR101 signaling facilitates LH release via indirect activation of kisspeptin neurons and that glutamatergic neurons may mediate the signaling. This provides a new aspect of kisspeptin- and GnRH-neuronal communication with the presence of stimulation from GnRH to kisspeptin neurons in female rats.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Hypothalamus, Anterior/drug effects , Kisspeptins/genetics , Luteinizing Hormone/drug effects , Neurons/metabolism , Peptide Fragments/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Female , Gene Knockout Techniques , Hypothalamus, Anterior/cytology , Hypothalamus, Anterior/metabolism , In Situ Hybridization , Injections, Intraventricular , Kisspeptins/pharmacology , Luteinizing Hormone/metabolism , Nerve Tissue Proteins/genetics , Ovariectomy , Rats , Rats, Transgenic , Receptors, G-Protein-Coupled/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Vesicular Glutamate Transport Protein 2/genetics
20.
Neuromolecular Med ; 20(4): 525-536, 2018 12.
Article in English | MEDLINE | ID: mdl-30218420

ABSTRACT

It is an established fact that orexin plays an important role in regulating the reproductive axis and the secretions of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH). However, its precise cellular and molecular mechanisms are not fully recognized. Accordingly, the aim of the present study is to find out whether the central injection of orexin A (OXA) and its antagonists, SB-334867 (as orexin receptor antagonist 1; OX1RA) and JNJ-10397049 (as orexin receptor antagonist 2; OX2RA), either alone or in combination, can leave any impact on the reproductive axis (either hormonal or behavioral) in the male Wistar rats. Furthermore, in order to see whether OXA signals can be relayed through the pathway of kisspeptin/neurokinin B/dynorphin (known as KNDy neurons, a neural network which works upstream of GnRH neurons) or not, the relative gene expression of these neuropeptides were measured. Overall, the data from radioimmunoassay revealed that OXA significantly decreases the mean serum level of LH and testosterone and, in a similar vein, its antagonists neutralize this impact. Moreover, data from real-time quantitative PCR indicated that OXA has significantly reduced the hypothalamic expression of Gnrh. In this line, the gene expressions of Kisspeptin and Neurokinin b decreased. However, OXA antagonists neutralize this impact. Also, the expression of Dynorphin gene was upregulated by the following application of the OXA. The results of this study are related to the impact of orexin on the reproductive axis. It is recommended that KNDy neurons as the interneural pathway relay the information of orexin to the GnRH neurons.


Subject(s)
Dynorphins/metabolism , Hypothalamus/drug effects , Kisspeptins/metabolism , Neurokinin B/metabolism , Neurons/drug effects , Orexins/pharmacology , Reproduction/drug effects , Animals , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacology , Dioxanes/administration & dosage , Dioxanes/pharmacology , Dynorphins/genetics , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/cytology , Injections, Intraventricular , Kisspeptins/genetics , Luteinizing Hormone/blood , Male , Naphthyridines , Neurokinin B/genetics , Neurons/metabolism , Orexins/administration & dosage , Orexins/antagonists & inhibitors , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , Random Allocation , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Sexual Behavior, Animal/drug effects , Testosterone/blood , Urea/administration & dosage , Urea/analogs & derivatives , Urea/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL