Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
1.
Biochem Biophys Res Commun ; 705: 149732, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38447390

RESUMEN

Neurokinin B (NKB) is a tachykinin peptide that has diverse roles in biology, including in human reproductive development. Cellular processing of this peptide is thought to involve formation of a dense core vesicle during transit through the regulated secretory pathway. The ability of NKB to rapidly form an amyloid can contribute to formation of the secretory granule but features that support amyloid formation of NKB are not well understood. NKB contains a diphenylalanine sequence well recognised as an important motif for self-assembly of other peptides including amyloid ß. Using mutations of the diphenylalanine motif we show that this motif in NKB is necessary for amyloid formation, and it is the unique combination of aromaticity and hydrophobicity of phenylalanine that is crucial for aggregation. Using disulfide cross-linking we propose that phenylalanine at sequence position 6 is important for stabilising inter-sheet interactions in the NKB amyloid fibril. Although having a highly conserved sequence, the NKB peptide from zebrafish only contains a single phenylalanine and does not fibrillise as extensively as mammalian NKB. Analysis of self-assembly of NKB-like peptides from different species may help in elucidating their biological roles. Taken together, this work shows that mammalian NKB has evolved, within only 10 residues, a sequence optimised for rapid self-assembly, whilst also containing residues for metal-binding, receptor binding and receptor discrimination.


Asunto(s)
Neuroquinina B , Neuropéptidos , Animales , Humanos , Neuroquinina B/química , Amiloide , Fenilalanina , Péptidos beta-Amiloides , Pez Cebra/metabolismo , Proteínas Amiloidogénicas , Mamíferos/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33500349

RESUMEN

The gonadotropin-releasing hormone (GnRH) pulse is fundamental for mammalian reproduction: GnRH pulse regimens are needed as therapies for infertile women as continuous GnRH treatment paradoxically inhibits gonadotropin release. Circumstantial evidence suggests that the hypothalamic arcuate KNDy neurons expressing kisspeptin (encoded by Kiss1), neurokinin B (encoded by Tac3), and dynorphin A serve as a GnRH pulse generator; however, no direct evidence is currently available. Here, we show that rescuing >20% KNDy neurons by transfecting Kiss1 inside arcuate Tac3 neurons, but not outside of these neurons, recovered folliculogenesis and luteinizing hormone (LH) pulses, an indicator of GnRH pulses, in female global Kiss1 knockout (KO) rats and that >90% conditional arcuate Kiss1 KO in newly generated Kiss1-floxed rats completely suppressed LH pulses. These results first provide direct evidence that KNDy neurons are the GnRH pulse generator, and at least 20% of KNDy neurons are sufficient to maintain folliculogenesis via generating GnRH/gonadotropin pulses.


Asunto(s)
Dinorfinas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Organogénesis , Folículo Ovárico/crecimiento & desarrollo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Aromatasa/genética , Aromatasa/metabolismo , Retroalimentación Fisiológica , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Integrasas/metabolismo , Hormona Luteinizante/sangre , Tamaño de los Órganos , Folículo Ovárico/metabolismo , Hipófisis/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de HL/genética , Receptores de HL/metabolismo , Receptores LHRH/metabolismo
3.
Front Neuroendocrinol ; 64: 100968, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34808231

RESUMEN

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.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Hormona Liberadora de Gonadotropina , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Dinorfinas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Mamíferos , Neuroquinina B/metabolismo , Neuronas/metabolismo
4.
Clin Endocrinol (Oxf) ; 98(6): 788-795, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36879296

RESUMEN

OBJECTIVE: There is controversial results about serum kisspeptin, neurokinin-B (NKB), anti-Müllerian hormone (AMH) and inhibin B (INHB) levels in girls with central precocious puberty (CPP). Aim of this study is to evaluate serum levels of these four peptides in patients presented with early pubertal signs, and to evaluate their diagnostic validity in the diagnosis of CPP. DESIGN: Cross-sectional study. PATIENTS: Study included 99 girls (51 CPP, 48 premature thelarche [PT]) whose breast development started before 8 years and 42 age-matched healthy prepubertal girls. Clinical findings, antropometric measurements, laboratory and radiological findings were recorded. Gonadotropin-releasing hormone (GnRH) stimulation test was performed in all cases with early breast development. MEASUREMENTS: Kisspeptin, NKB, INHB and AMH levels were measured in fasting serum samples using enzyme-linked immunosorbent assay method. RESULTS: There was no statistically significant difference between mean ages of girls with CPP (7.1 ± 1.2 years), PT (7.2 ± 1.3 years) and prepubertal controls (7.0 ± 1.0 years). Serum kisspeptin, NKB and INHB levels were higher in CPP group compared to PT and control groups, while serum AMH level was lower in CPP group. Serum kisspeptin, NKB, and INHB were all positively correlated with bone age (BA) advancement, and peak luteinizing hormone in GnRH test. Multiple stepwise regression analysis revealed that the most important factors used to differentiate CPP from PT were advanced BA, serum kisspeptin, NKB and INHB levels (AUC: 0.819, p < .001). CONCLUSIONS: We, first showed in the same patients' group that serum kisspeptin, NKB and INHB were higher in patients with CPP and can be used as alternative parameters to distinguish CPP from PT.


Asunto(s)
Kisspeptinas , Pubertad Precoz , Femenino , Humanos , Preescolar , Niño , Pubertad Precoz/diagnóstico , Neuroquinina B , Estudios Transversales , Pubertad , Hormona Antimülleriana , Hormona Liberadora de Gonadotropina , Hormona Folículo Estimulante
5.
J Reprod Dev ; 69(4): 218-222, 2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37271516

RESUMEN

Secretion of pulsatile gonadotropin-releasing hormone (GnRH) is essential for reproduction. Kisspeptin neurons in the arcuate nucleus (ARC), which coexpress neurokinin B (NKB) and its receptor (NK3R), are believed to be components of the GnRH pulse generator that regulates pulsatile GnRH secretion. We examined the effects of peripheral infusion of senktide, an NK3R selective agonist, on GnRH pulse generator activity by monitoring multiple unit activity (MUA) in the goat ARC. Previous studies have shown that characteristic increases in MUA (MUA volleys) reflect GnRH pulse generator activity. Senktide was infused intravenously or intravaginally for 2 h while recording MUA. Both infusions significantly increased the MUA volley frequency compared with the control. These results demonstrate that peripherally administered senktide acts centrally to sustainably accelerate the neural activity of the GnRH pulse generator throughout the infusion period. This suggests the possibility of practical applications of NK3R agonists for improving reproductive activity in farm animals.


Asunto(s)
Hormona Liberadora de Gonadotropina , Receptores de Neuroquinina-3 , Animales , Receptores de Neuroquinina-3/agonistas , Hormona Luteinizante , Cabras , Hormonas Esteroides Gonadales , Neuroquinina B , Kisspeptinas/farmacología
6.
Gynecol Endocrinol ; 39(1): 2216313, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37224872

RESUMEN

BACKGROUND: Neurokinin B (NKB) belongs to the tachykinin family of proteins who's regulation is essential for proper function of the reproductive system. It has been shown that patients with functional hypothalamic amenorrhea (FHA) exhibit decreased levels of serum kisspeptin. As kisspeptin secretion is regulated by NKB signaling, it is reasonable to suspect that patients with FHA will also have abnormal NKB secretion. AIM: To assess NKB levels in patients with FHA and to determine whether NKB signaling is affected in these patients. We hypothesized that decreased NKB signaling is a factor contributing to the development of the FHA. MATERIALS AND METHODS: A total of 147 patients with FHA and 88 healthy age-matched controls were enrolled. Baseline blood samples were drawn from both groups to measure serum concentrations of NKB, luteinizing hormone (LH), follicle-stimulating hormone (FSH), estradiol (E2), prolactin (PRL), thyroid-stimulating hormone (TSH), free thyroxine (fT4), cortisol, dehydroepiandrosterone sulfate (DHEA-S), testosterone (T), glucose, and insulin. RESULTS: Mean serum NKB levels were found to be decreased significantly in the FHA group when compared with the control group (628.35 ± 324.92 vs. 721.41 ± 337.57 ng/L, respectively p = 0.002). No statistical difference was observed in NKB-1 levels within the FHA group when selecting for normal and decreased body mass index. CONCLUSIONS: Patients with FHA were found to have decreased serum NKB concentrations when compared to healthy controls. Abnormal NKB secretion is likely a key factor contributing to development of FHA.


Asunto(s)
Amenorrea , Neuroquinina B , Femenino , Humanos , Amenorrea/etiología , Kisspeptinas , Factores de Riesgo , Estradiol
7.
Molecules ; 28(18)2023 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-37764220

RESUMEN

Amyloid diseases have become a global concern due to their increasing prevalence. Transition metals, including copper, can affect the aggregation of the pathological proteins involved in these diseases. Copper ions play vital roles in organisms, but the disruption of their homeostasis can negatively impact neuronal function and contribute to amyloid diseases with toxic protein aggregates, oxidative stress, mitochondrial dysfunction, impaired cellular signaling, inflammation, and cell death. Gaining insight into the imbalance of copper ions and its impact on protein folding and aggregation is crucial for developing focused therapies. This review examines the influence of copper ions on significant amyloid proteins/peptides, offering a comprehensive overview of the current understanding in this field.


Asunto(s)
Enfermedad de Alzheimer , Proteínas Amiloidogénicas , Humanos , Cobre/metabolismo , Pliegue de Proteína , Agregado de Proteínas , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo
8.
J Pharm Technol ; 39(6): 291-297, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37974591

RESUMEN

Objective: To review the safety, efficacy, and tolerability of fezolinetant for the treatment of vasomotor symptoms associated with menopause. Data Sources: A literature search was conducted through PubMed using the following search terms: fezolinetant, ES259564, SKYLIGHT, vasomotor symptoms, and menopause. Study Selection and Data Extraction: Selected articles included those which described clinical studies of the pharmacology, pharmacokinetics, efficacy, safety, or tolerability of fezolinetant. Data Synthesis: Fezolinetant works by inhibiting neurokinin B from binding to its receptor in the hypothalamus, thereby decreasing the occurrence of vasomotor symptoms. Clinical trials have demonstrated fezolinetant is superior to placebo in decreasing the frequency and severity of vasomotor symptoms. Common adverse effects associated with the use of fezolinetant include headache and gastrointestinal disturbances, as well as elevations in liver transaminase levels. Conclusions: Clinical practice guidelines for the treatment of vasomotor symptoms associated with menopause recommend hormone therapy as the most effective treatment option. Risks associated with hormone use may limit the use of this option in some patients. Neurokinin B inhibitors, like fezolinetant, target the physiologic cause of vasomotor symptoms. With the approval of fezolinetant, as well as elinzanetant which is currently in phase 3 clinical trials, providers and patients have additional nonhormonal treatment options for vasomotor symptoms associated with menopause.

9.
J Neurosci ; 41(47): 9688-9701, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34654752

RESUMEN

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).


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/fisiología , Kisspeptinas/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Animales , Señalización del Calcio/fisiología , Dieta Alta en Grasa , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Proteínas Fluorescentes Verdes , Ratones
10.
J Neurosci ; 41(46): 9539-9560, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34642212

RESUMEN

The lateral hypothalamic area (LHA) is a highly conserved brain region critical for maintaining physiological homeostasis and goal-directed behavior. LHA neurons that express melanin-concentrating hormone (MCH) are key regulators of arousal, energy balance, and motivated behavior. However, cellular and functional diversity among LHAMCH neurons is not well understood. Previous anatomic and molecular data suggest that LHAMCH neurons may be parsed into at least two distinct subpopulations, one of which is enriched in neurokinin-3 receptor (NK3R), the receptor for neurokinin B (NKB), encoded by the Tac2 gene. This tachykininergic ligand-receptor system has been implicated in reproduction, fear memory, and stress in other brain regions, but NKB interactions with LHAMCH neurons are poorly understood. We first identified how LHAMCH subpopulations may be distinguished anatomically and electrophysiologically. To dissect functional connectivity between NKB-expressing neurons and LHAMCH neurons, we used Cre-dependent retrograde and anterograde viral tracing in male Tac2-Cre mice and identified Tac2/EYFP+ neurons in the bed nucleus of the stria terminalis and central nucleus of the amygdala, the central extended amygdala, as major sources of NKB input onto LHAMCH neurons. In addition to innervating the LHA, these limbic forebrain NKB neurons also project to midbrain and brainstem targets. Finally, using a dual-virus approach, we found that optogenetic activation of these inputs in slices evokes GABA release onto a subset of LHAMCH neurons but lacked specificity for the NK3R+ subpopulation. Overall, these data define parallel tachykininergic/GABAergic limbic forebrain projections that are positioned to modulate multiple nodes of homeostatic and behavioral control.SIGNIFICANCE STATEMENT The LHA orchestrates fundamental behavioral states in the mammalian hypothalamus, including arousal, energy balance, memory, stress, and motivated behavior. The neuropeptide MCH defines one prominent population of LHA neurons, with multiple roles in the regulation of homeostatic behavior. Outstanding questions remain concerning the upstream inputs that control MCH neurons. We sought to define neurochemically distinct pathways in the mouse brain that may communicate with specific MCH neuron subpopulations using viral-based retrograde and anterograde neural pathway tracing and optogenetics in brain slices. Here, we identify a specific neuropeptide-defined forebrain circuit that makes functional synaptic connections with MCH neuron subpopulations. This work lays the foundation for further manipulating molecularly distinct neural circuits that modulate innate behavioral states.


Asunto(s)
Núcleo Amigdalino Central/citología , Área Hipotalámica Lateral/citología , Vías Nerviosas/citología , Neuronas/citología , Animales , Hormonas Hipotalámicas/metabolismo , Masculino , Melaninas/metabolismo , Ratones , Ratones Transgénicos , Vías Nerviosas/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo
11.
Clin Endocrinol (Oxf) ; 97(2): 156-164, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35262967

RESUMEN

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age and is the leading cause of anovulatory subfertility. Increased gonadotrophin releasing hormone (GnRH) pulsatility in the hypothalamus results in preferential luteinizing hormone (LH) secretion from the pituitary gland, leading to ovarian hyperandrogenism and oligo/anovulation. The resultant hyperandrogenism reduces negative feedback from sex steroids such as oestradiol and progesterone to the hypothalamus, and thus perpetuates the increase in GnRH pulsatility. GnRH neurons do not have receptors for oestrogen, progesterone, or androgens, and thus the disrupted feedback is hypothesized to occur via upstream neurons. Likely candidates for these upstream regulators of GnRH neuronal pulsatility are Kisspeptin, Neurokinin B (NKB), and Dynorphin neurons (termed KNDy neurons). Growing insight into the neuroendocrine dysfunction underpinning the heightened GnRH pulsatility seen in PCOS has led to research on the use of pharmaceutical agents that specifically target the activity of these KNDy neurons to attenuate symptoms of PCOS. This review aims to highlight the neuroendocrine abnormalities that lead to increased GnRH pulsatility in PCOS, and outline data on recent therapeutic advancements that could potentially be used to treat PCOS. Emerging evidence has investigated the use of neurokinin 3 receptor (NK3R) antagonists as a method of reducing GnRH pulsatility and alleviating features of PCOS such as hyperandrogenism. We also consider other potential mechanisms by which increased GnRH pulsatility is controlled, which could form the basis of future avenues of research.


Asunto(s)
Hiperandrogenismo , Síndrome del Ovario Poliquístico , Femenino , Hormona Liberadora de Gonadotropina , Humanos , Hiperandrogenismo/tratamiento farmacológico , Kisspeptinas , Hormona Luteinizante , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Progesterona
12.
Int Immunol ; 33(12): 731-736, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34491348

RESUMEN

Atopic dermatitis (AD) is one of the most prevalent chronic inflammatory skin diseases in the world. It is characterized by recurrent eczematous lesions and intense itch, and many cytokines are involved in the pathogenesis of AD. Among them, much attention has been paid to interleukin 31 (IL-31) as an AD-associated itch mediator. IL-31 is mainly produced by CD4+ helper T cells and transmits the signals via a heterodimeric receptor composed of IL-31 receptor A (IL-31RA) and oncostatin M receptor (OSMR), both of which are expressed in dorsal root ganglion (DRG) neurons. However, the molecular mechanisms of how IL-31 is produced in helper T cells upon stimulation and transmits the itch sensation to the brain were largely unknown. Recently, by using original mouse models of AD, we have identified endothelial PAS domain 1 (EPAS1) and neurokinin B (NKB) as key molecules critical for IL-31 production and IL-31-mediated itch transmission, respectively. These molecules could be novel drug targets for AD-associated itch. This review highlights our recent findings, which show the functional significance of these molecules in the IL-31-induced itch sensation, referring to their application to drug development.


Asunto(s)
Desarrollo de Medicamentos , Interleucinas/inmunología , Animales , Dermatitis Atópica , Humanos , Interleucinas/biosíntesis
13.
Gen Comp Endocrinol ; 317: 113974, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34973969

RESUMEN

Kisspeptin (KISS), a key hormone involved in the regulation of the hypothalamic-pituitary-ovarian (HPO) axis, has been localized in the anteroventral periventricular (AVPV) nucleus and the neighboring rostral periventricular nucleus (PeVN), and in the arcuate (ARC) nucleus of the mammalian hypothalamus. In the ARC, the KISS neurons that co-express neurokinin B (NKB) and dynorphin A (Dyn) are named KNDy cells. The South American plains vizcacha is a rodent with peculiar reproductive traits. Around mid-pregnancy, vizcacha shows the reactivation of its HPO axis with the pulsatile release of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH), an essential event for the success of gestation. Considering the role of KISS system in GnRH modulation, the aim of this work was to study their neuroanatomical distribution in adult vizcachas. AVPV showed sexual dimorphism with a significant smaller area in males (t-Test, p < 0.05), and KISS immunoreactivity was detected in somas and varicosities homogenously distributed in the AVPV with a concordant sex-related expression pattern. NKB and Dyn expression was also observed in cytoplasm of neurons scattered in the AVPV. Three subpopulations of neurons were detected in the AVPV: neurons expressing Dyn and NKB (DyNK cells), neurons expressing KISS and NKB (KiNK cells), and single NKB expressing neurons. Strikingly, KISS and Dyn were always expressed in different cells. In addition, in the ARC nucleus, KNDy cells were detected. On the other hand, KISS and GnRH expression was detected in different subpopulations of neurons, GnRH cells showed KISS receptor (KISSR or GPR-54) expression, and KISS immunoreactive afferent contacts were detected making close appositions onto somas and dendrites of GnRH cells. These results show similarities and differences between the KISS system in the hypothalamus of the vizcacha and other mammals, and constitute crucial observations about KISS and GnRH relation. Considering the peculiarity of HPO axis regulation in this species, the present work provides a neuroanatomical framework for the further elucidation of molecular mechanisms underlying GnRH expression and secretion.


Asunto(s)
Hormona Liberadora de Gonadotropina , Kisspeptinas , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Masculino , Mamíferos/metabolismo , Neuroquinina B/metabolismo , Embarazo , América del Sur
14.
Gynecol Endocrinol ; 38(11): 997-1002, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36170883

RESUMEN

Background: Functional hypothalamic amenorrhea (FHA) is a chronic endocrine disorder caused by the abnormal pulsatile secretion of neurohormones in the hypothalamus. Secretion of GnRH is regulated by kisspeptin/neurokinin B/dynorphin (KNDy) neurons. These neurons produce, among other neurohormones, neurokinin B (NKB) which regulates the coordinated stimulation or inhibition of GnRH secreting neurons. Aim of the study: Assessment and comparison of serum NKB in patients with FHA at baseline, and following 6 months of estrogen-progestagen therapy. Materials and methods: Fifty-five patients with functional hypothalamic amenorrhea were included in the study group. Serum concentrations of neurokinin B (NKB), follicle stimulating hormone (FSH), luteinizing hormone (LH), 17-ß-estradiol (E2), prolactin (PRL), cortisol, testosterone (T), dehydroepiandrosterone sulfate (DHEA-S), thyroid-stimulating hormone (TSH), free thyroxine (fT4), fasting glucose and insulin, as well as lipid profile were measured at baseline. At the time of diagnosis, patients with FHA were prescribed a course of 2 mg 17-ß-estradiol and 10 mg dydrogesterone for duration of 6 months. Serum NKB was then reassessed following treatment at 6 months. Results: At baseline, the FHA group was found to have a decreased serum NKB concentration when compared to a healthy control group. Following 6 months of sequential estrogen-progestogen hormone therapy, this study did not find any statistically significant difference in serum NKB concentration in the treatment arm compared to baseline. Conclusions: For the first time, NKB secretion has been studied in patients with FHA. A significantly lower level of serum NKB was observed in these patients at baseline, when compared to a control group. After 6 months of combination estrogen-progesterone therapy, no significant changes in NKB levels were observed in these patients. These findings, for the first time in the literature, provide insight into the perceived benefit of HRT, calling into question its benefit in addressing the underlying etiopathogenetic contributors of FHA. These new findings may contribute to more targeted and appropriate treatment of such patients in the future.


Asunto(s)
Neuroquinina B , Progestinas , Femenino , Humanos , Progestinas/uso terapéutico , Amenorrea , Hormona Liberadora de Gonadotropina , Estrógenos , Estradiol , Neurotransmisores , Kisspeptinas
15.
Biol Reprod ; 105(4): 1056-1067, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34037695

RESUMEN

Mechanisms in the brain controlling secretion of gonadotropin hormones in pigs, particularly luteinizing hormone (LH), are poorly understood. Kisspeptin is a potent LH stimulant that is essential for fertility in many species, including pigs. Neurokinin B (NKB) acting through neurokinin 3 receptor (NK3R) is involved in kisspeptin-stimulated LH release, but organization of NKB and NK3R within the porcine hypothalamus is unknown. Hypothalamic tissue from ovariectomized (OVX) gilts was used to determine the distribution of immunoreactive kisspeptin, NKB, and NK3R cells in the arcuate nucleus (ARC). Almost all kisspeptin neurons coexpressed NKB in the porcine ARC. Immunostaining for NK3R was distributed throughout the preoptic area (POA) and in several hypothalamic areas including the periventricular and retrochiasmatic areas but was not detected within the ARC. There was no colocalization of NK3R with gonadotropin-releasing hormone (GnRH), but NK3R-positive fibers in the POA were in close apposition to GnRH neurons. Treating OVX gilts with the progestin altrenogest decreased LH pulse frequency and reduced mean circulating concentrations of LH compared with OVX control gilts (P < 0.01), but the number of kisspeptin and NKB cells in the ARC did not differ between treatments. The neuroanatomical arrangement of kisspeptin, NKB, and NK3R within the porcine hypothalamus confirms they are positioned to stimulate GnRH and LH secretion in gilts, though differences with other species exist. Altrenogest suppression of LH secretion in the OVX gilt does not appear to involve decreased peptide expression of kisspeptin or NKB.


Asunto(s)
Hipotálamo/metabolismo , Kisspeptinas/genética , Neuroquinina B/genética , Progestinas/farmacología , Receptores de Neuroquinina-3/genética , Sus scrofa/genética , Acetato de Trembolona/análogos & derivados , Animales , Femenino , Perfilación de la Expresión Génica/veterinaria , Hipotálamo/efectos de los fármacos , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Receptores de Neuroquinina-3/metabolismo , Sus scrofa/metabolismo , Acetato de Trembolona/farmacología
16.
Neuroendocrinology ; 111(12): 1151-1163, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33406521

RESUMEN

The pubertal process is initiated as a result of complex neuroendocrine interactions within the preoptic and hypothalamic regions of the brain. These interactions ultimately result in a timely increase in the secretion of gonadotropin-releasing hormone (GnRH). Researchers for years have believed that this increase is due to a diminished inhibitory tone which has applied a prepubertal brake on GnRH secretion, as well as to the gradual development of excitatory inputs driving the increased release of the peptide. Over the years, insulin-like growth factor-1 (IGF-1) has emerged as a prime candidate for playing an important role in the onset of puberty. This review will first present initial research demonstrating that IGF-1 increases in circulation as puberty approaches, is able to induce the release of prepubertal GnRH, and can advance the timing of puberty. More recent findings depict an early action of IGF-1 to activate a pathway that releases the inhibitory brake on prepubertal GnRH secretion provided by dynorphin, as well as demonstrating that IGF-1 can also act later in the process to regulate the synthesis and release of kisspeptin, a potent stimulator of GnRH at puberty.


Asunto(s)
Dinorfinas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Kisspeptinas/metabolismo , Pubertad/metabolismo , Animales , Humanos
17.
J Reprod Dev ; 67(6): 352-358, 2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34629331

RESUMEN

Kisspeptin neurons in the arcuate nucleus (ARC), which co-express neurokinin B (NKB) and dynorphin A, are termed KNDy neurons. These neurons are candidates for the intrinsic gonadotropin-releasing hormone (GnRH) pulse generator. The central and peripheral administration of NKB or its receptor (NK3R) agonist evokes GnRH pulse generator activity and the subsequent pulsatile GnRH/luteinizing hormone (LH) secretion. However, the mechanism responsible for neural activation of the GnRH pulse generator in goats is unclear. We conducted electrophysiological and histochemical experiments to test the hypothesis that KNDy neurons receive NKB and that the signal is transmitted bilaterally to a population of KNDy neurons. Bilateral electrodes aimed at a cluster of KNDy neurons were inserted into the ovariectomized goat ARC. We observed the GnRH pulse generator activity, represented by characteristic increases in multiple-unit activity (MUA volleys). The unilateral administration of NKB or vehicle in the close vicinity of KNDy neurons under simultaneous MUA recording from both sides revealed that only NKB evoked MUA volley(s) immediately after administration. The timing of the MUA volley(s) evoked on the ipsilateral side was synchronized to that on the contralateral side. The double-labeled ISH for KISS1 and TACR3, which encode kisspeptin and NK3R, respectively, revealed that most KNDy neurons co-expressed TACR3. Therefore, NKB could directly stimulate KNDy neurons, following which the stimulatory signal is immediately transmitted to the entire population of KNDy neurons via connection with their fibers. This mechanism helps synchronize burst activity among KNDy neurons, thereby generating neural signals that govern pulsatile GnRH secretion.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Neuroquinina B , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Dinorfinas/metabolismo , Cabras , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo
18.
Endocr J ; 68(8): 933-941, 2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-33867395

RESUMEN

The brain mechanism responsible for the pulsatile secretion of gonadotropin-releasing hormone (GnRH) is important for maintaining reproductive function in mammals. Accumulating evidence suggests that kisspeptin/neurokinin B/dynorphin A (KNDy) neurons in the hypothalamic arcuate nucleus (ARC) play a critical role in the regulation of pulsatile GnRH and subsequent gonadotropin secretion. Dynorphin A (Dyn) and its receptor, kappa-opioid receptor (KOR, encoded by Oprk1), have been shown to be involved in the suppression of pulsatile GnRH/luteinizing hormone (LH) release. On the other hand, it is still unclear whether the inhibitory Dyn signaling affects KNDy neurons or KOR-expressing non-KNDy cells in the ARC or other brain regions. We therefore aimed to clarify the role of ARC-specific Dyn-KOR signaling in the regulation of pulsatile GnRH/LH release by the ARC specific cell deletion of KOR-expressing cells using Dyn-conjugated-saporin (Dyn-SAP). Estrogen-primed ovariectomized female rats were administered Dyn-SAP to the ARC. In situ hybridization of Oprk1 showed that ARC Dyn-SAP administration significantly decreased the number of Oprk1-expressing cells in the ARC, but not in the ventromedial hypothalamic nucleus and paraventricular nucleus. The frequency of LH pulses significantly increased in animals bearing the ARC Dyn-SAP administration. The number of Kiss1-expressing cells in the ARC was not affected by ARC Dyn-SAP treatment. Dyn-KOR signaling within the ARC seems to mediate the suppression of the frequency of pulsatile GnRH/LH release, and ARC non-KNDy KOR neurons may be involved in the mechanism modulating GnRH/LH pulse generation.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Neuronas/metabolismo , Receptores Opioides kappa/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Dinorfinas/administración & dosificación , Femenino , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Saponinas/administración & dosificación
19.
Int J Neurosci ; 131(8): 780-788, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32303141

RESUMEN

AIMS: A number of studies have shown that neuropeptide Y (NPY) is considered to be one of the key regulators of hypothalamic-pituitary-gonadal (HPG) axis in the mammals. In addition, kisspeptin (encode by Kiss1 gene), neurokinin B (encode by Tac3 gene) and dynorphin (encode by Pdyn gene) (commonly known as KNDy secreting neurons) are a powerful upstream regulators of GnRH neuron in hypothalamus. MATERIALS AND METHODS: The present study aims to investigate the effects of the intracerebroventricular (icv) injection of NPY and BIBP3226 (NPY receptor antagonist (NPYRA)) on the male sexual behavioral. Additionally, in order to see whether NPY signals can be relayed through the pathway of kisspeptin/neurokinin B/dynorphin, the gene expression of these peptides along with Gnrh1 gene in the hypothalamus were measured. RESULTS: The icv injection of NPY decreased the latencies and increase the frequencies of sexual parameters of the male rats in a significant way. In this line, NPYRA antagonized the stimulative effects of NPY. Moreover, data from real-time quantitative PCR indicated that injection of NPY significantly increased the gene expression of Gnrh1, Kiss1 and Tac3 and decrease the Pdyn while treatment with NPYRA controlled the modulative effects of NPY on these gene expression. CONCLUSIONS: In conclusion based on the results of this study, NPY can exert its impacts on the sexual behavior of male rats via modulation of the KNDy secreting neurons as an interneural pathway to GnRH neurons.


Asunto(s)
Neuropéptido Y/administración & dosificación , Neuropéptido Y/fisiología , Conducta Sexual Animal/efectos de los fármacos , Conducta Sexual Animal/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Dinorfinas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Kisspeptinas , Masculino , Neuroquinina B/metabolismo , Ratas Wistar
20.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34502135

RESUMEN

Estrogen produced by ovarian follicles plays a key role in the central mechanisms controlling reproduction via regulation of gonadotropin-releasing hormone (GnRH) release by its negative and positive feedback actions in female mammals. It has been well accepted that estrogen receptor α (ERα) mediates both estrogen feedback actions, but precise targets had remained as a mystery for decades. Ever since the discovery of kisspeptin neurons as afferent ERα-expressing neurons to govern GnRH neurons, the mechanisms mediating estrogen feedback are gradually being unraveled. The present article overviews the role of kisspeptin neurons in the arcuate nucleus (ARC), which are considered to drive pulsatile GnRH/gonadotropin release and folliculogenesis, in mediating the estrogen negative feedback action, and the role of kisspeptin neurons located in the anteroventral periventricular nucleus-periventricular nucleus (AVPV-PeN), which are thought to drive GnRH/luteinizing hormone (LH) surge and consequent ovulation, in mediating the estrogen positive feedback action. This implication has been confirmed by the studies showing that estrogen-bound ERα down- and up-regulates kisspeptin gene (Kiss1) expression in the ARC and AVPV-PeN kisspeptin neurons, respectively. The article also provides the molecular and epigenetic mechanisms regulating Kiss1 expression in kisspeptin neurons by estrogen. Further, afferent ERα-expressing neurons that may regulate kisspeptin release are discussed.


Asunto(s)
Encéfalo/metabolismo , Estrógenos/metabolismo , Retroalimentación Fisiológica , Kisspeptinas/metabolismo , Ovulación , Animales , Encéfalo/citología , Encéfalo/fisiología , Femenino , Humanos , Neuronas/metabolismo , Neuronas/fisiología , Receptores de Estrógenos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda