Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 15: 1353151, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38348415

RESUMEN

Reproduction in mammals is controlled by hypothalamic gonadotropin-releasing hormone (GnRH) neurons. Recent studies from our laboratory established that the basal ganglia of the human brain contain additional large populations of GnRH synthesizing neurons which are absent in adult mice. Such extrahypothalamic GnRH neurons mostly occur in the putamen where they correspond to subsets of the striatal cholinergic interneurons (ChINs) and express GnRHR autoreceptors. In an effort to establish a mouse model for functional studies of striatal GnRH/GnRHR signaling, we carried out electrophysiological experiments on acute brain slices from male transgenic mice. Using PN4-7 neonatal mice, half of striatal ChINs responded with transient hyperpolarization and decreased firing rate to 1.2 µM GnRH, whereas medium spiny projection neurons remained unaffected. GnRH acted on its specific receptor because no response was observed in the presence of the GnRHR antagonist Antide. Addition of the membrane-impermeable G protein-coupled receptor inhibitor GDP-ß-S to the internal electrode solution eliminated the effect of GnRH. Further, GnRH was able to inhibit ChINs in presence of tetrodotoxin which blocked action potential mediated events. Collectively, these data indicated that the receptor underlying the effects of GnRH in neonatal mice is localized within ChINs. GnRH responsiveness of ChINs was transient and entirely disappeared in adult mice. These results raise the possibility to use neonatal transgenic mice as a functional model to investigate the role of GnRH/GnRHR signaling discovered earlier in adult human ChINs.


Asunto(s)
Hormona Liberadora de Gonadotropina , Receptores LHRH , Animales , Masculino , Ratones , Neuronas Colinérgicas , Hormona Liberadora de Gonadotropina/farmacología , Mamíferos , Ratones Transgénicos , Transducción de Señal
2.
Front Endocrinol (Lausanne) ; 13: 960769, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36093104

RESUMEN

Kisspeptin neurons residing in the rostral periventricular area of the third ventricle (KPRP3V) and the arcuate nucleus (KPARC) mediate positive and negative estrogen feedback, respectively. Here, we aim to compare transcriptional responses of KPRP3V and KPARC neurons to estrogen. Transgenic mice were ovariectomized and supplemented with either 17ß-estradiol (E2) or vehicle. Fluorescently tagged KPRP3V neurons collected by laser-capture microdissection were subjected to RNA-seq. Bioinformatics identified 222 E2-dependent genes. Four genes encoding neuropeptide precursors (Nmb, Kiss1, Nts, Penk) were robustly, and Cartpt was subsignificantly upregulated, suggesting putative contribution of multiple neuropeptides to estrogen feedback mechanisms. Using overrepresentation analysis, the most affected KEGG pathways were neuroactive ligand-receptor interaction and dopaminergic synapse. Next, we re-analyzed our previously obtained KPARC neuron RNA-seq data from the same animals using identical bioinformatic criteria. The identified 1583 E2-induced changes included suppression of many neuropeptide precursors, granins, protein processing enzymes, and other genes related to the secretory pathway. In addition to distinct regulatory responses, KPRP3V and KPARC neurons exhibited sixty-two common changes in genes encoding three hormone receptors (Ghsr, Pgr, Npr2), GAD-65 (Gad2), calmodulin and its regulator (Calm1, Pcp4), among others. Thirty-four oppositely regulated genes (Kiss1, Vgf, Chrna7, Tmem35a) were also identified. The strikingly different transcriptional responses in the two neuron populations prompted us to explore the transcriptional mechanism further. We identified ten E2-dependent transcription factors in KPRP3V and seventy in KPARC neurons. While none of the ten transcription factors interacted with estrogen receptor-α, eight of the seventy did. We propose that an intricate, multi-layered transcriptional mechanism exists in KPARC neurons and a less complex one in KPRP3V neurons. These results shed new light on the complexity of estrogen-dependent regulatory mechanisms acting in the two functionally distinct kisspeptin neuron populations and implicate additional neuropeptides and mechanisms in estrogen feedback.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Kisspeptinas , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Estrógenos/metabolismo , Estrógenos/farmacología , Kisspeptinas/genética , Kisspeptinas/metabolismo , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Factores de Transcripción/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(27): e2113749119, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35763574

RESUMEN

Kisspeptin neurons in the mediobasal hypothalamus (MBH) are critical targets of ovarian estrogen feedback regulating mammalian fertility. To reveal molecular mechanisms underlying this signaling, we thoroughly characterized the estrogen-regulated transcriptome of kisspeptin cells from ovariectomized transgenic mice substituted with 17ß-estradiol or vehicle. MBH kisspeptin neurons were harvested using laser-capture microdissection, pooled, and subjected to RNA sequencing. Estrogen treatment significantly (p.adj. < 0.05) up-regulated 1,190 and down-regulated 1,139 transcripts, including transcription factors, neuropeptides, ribosomal and mitochondrial proteins, ion channels, transporters, receptors, and regulatory RNAs. Reduced expression of the excitatory serotonin receptor-4 transcript (Htr4) diminished kisspeptin neuron responsiveness to serotonergic stimulation. Many estrogen-regulated transcripts have been implicated in puberty/fertility disorders. Patients (n = 337) with congenital hypogonadotropic hypogonadism (CHH) showed enrichment of rare variants in putative CHH-candidate genes (e.g., LRP1B, CACNA1G, FNDC3A). Comprehensive characterization of the estrogen-dependent kisspeptin neuron transcriptome sheds light on the molecular mechanisms of ovary-brain communication and informs genetic research on human fertility disorders.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Estrógenos , Fertilidad , Kisspeptinas , Neuronas , Ovario , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Estrógenos/metabolismo , Femenino , Fertilidad/genética , Perfilación de la Expresión Génica , Humanos , Hipogonadismo/congénito , Hipogonadismo/genética , Kisspeptinas/genética , Kisspeptinas/metabolismo , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Ovario/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-36910428

RESUMEN

Immunization is one of the most effective public health interventions, saving millions of lives every year. Ethiopia has seen gradual improvements in immunization coverage and access to child health care services; however, inequalities in child mortality across wealth quintiles and regions remain persistent. We model the relative distributional incidence and mortality of four vaccine-preventable diseases (VPDs) (rotavirus diarrhea, human papillomavirus, measles, and pneumonia) by wealth quintile and geographic region in Ethiopia. Our approach significantly extends an earlier methodology, which utilizes the population attributable fraction and differences in the prevalence of risk and prognostic factors by population subgroup to estimate the relative distribution of VPD incidence and mortality. We use a linear system of equations to estimate the joint distribution of risk and prognostic factors in population subgroups, treating each possible combination of risk or prognostic factors as computationally distinct, thereby allowing us to account for individuals with multiple risk factors. Across all modeling scenarios, our analysis found that the poor and those living in rural and primarily pastoralist or agrarian regions have a greater risk than the rich and those living in urban regions of becoming infected with or dying from a VPD. While in absolute terms all population subgroups benefit from health interventions (e.g., vaccination and treatment), current unequal levels and pro-rich gradients of vaccination and treatment-seeking patterns should be redressed so to significantly improve health equity across wealth quintiles and geographic regions in Ethiopia.

5.
Handb Clin Neurol ; 180: 275-296, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34225935

RESUMEN

In mammals, kisspeptin neurons are the key components of the hypothalamic neuronal networks that regulate the onset of puberty, account for the pulsatile secretion of gonadotropin-releasing hormone (GnRH) and mediate negative and positive estrogen feedback signals to GnRH neurons. Being directly connected anatomically and functionally to the hypophysiotropic GnRH system, the major kisspeptin cell groups of the preoptic area/rostral hypothalamus and the arcuate (or infundibular) nucleus, respectively, are ideally positioned to serve as key nodes which integrate various types of environmental, endocrine, and metabolic signals that can influence fertility. This chapter provides an overview of the current state of knowledge on the anatomy, functions, and plasticity of brain kisspeptin systems based on the wide literature available from different laboratory and domestic species. Then, the species-specific features of human hypothalamic kisspeptin neurons are described, covering their topography, morphology, unique neuropeptide content, plasticity, and connectivity to hypophysiotropic GnRH neurons. Some newly emerging roles of central kisspeptin signaling in behavior and finally, clinical perspectives, are discussed.


Asunto(s)
Kisspeptinas , Neuroanatomía , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Kisspeptinas/metabolismo
6.
Elife ; 102021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128468

RESUMEN

Human reproduction is controlled by ~2000 hypothalamic gonadotropin-releasing hormone (GnRH) neurons. Here, we report the discovery and characterization of additional ~150,000-200,000 GnRH-synthesizing cells in the human basal ganglia and basal forebrain. Nearly all extrahypothalamic GnRH neurons expressed the cholinergic marker enzyme choline acetyltransferase. Similarly, hypothalamic GnRH neurons were also cholinergic both in embryonic and adult human brains. Whole-transcriptome analysis of cholinergic interneurons and medium spiny projection neurons laser-microdissected from the human putamen showed selective expression of GNRH1 and GNRHR1 autoreceptors in the cholinergic cell population and uncovered the detailed transcriptome profile and molecular connectome of these two cell types. Higher-order non-reproductive functions regulated by GnRH under physiological conditions in the human basal ganglia and basal forebrain require clarification. The role and changes of GnRH/GnRHR1 signaling in neurodegenerative disorders affecting cholinergic neurocircuitries, including Parkinson's and Alzheimer's diseases, need to be explored.


Asunto(s)
Ganglios Basales , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas , Adulto , Prosencéfalo Basal/citología , Ganglios Basales/citología , Ganglios Basales/metabolismo , Ganglios Basales/fisiología , Células Cultivadas , Colina O-Acetiltransferasa , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/citología , Neuronas/metabolismo , Neuronas/fisiología , Putamen/citología , Transcriptoma
7.
Neuroendocrinology ; 111(3): 249-262, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32299085

RESUMEN

BACKGROUND: Kisspeptin (KP) neurons in the rostral periventricular region of the 3rd ventricle (RP3V) of female rodents mediate positive estrogen feedback to gonadotropin-releasing hormone neurons and, thus, play a fundamental role in the mid-cycle luteinizing hormone (LH) surge. The RP3V is sexually dimorphic, and male rodents with lower KP cell numbers are unable to mount estrogen-induced LH surges. OBJECTIVE: To find and characterize the homologous KP neurons in the human brain, we studied formalin-fixed post-mortem hypothalami. METHODS: Immunohistochemical techniques were used. RESULTS: The distribution of KP neurons in the rostral hypothalamus overlapped with distinct subdivisions of the paraventricular nucleus. The cell numbers decreased after menopause, indicating that estrogens positively regulate KP gene expression in the rostral hypothalamus in humans, similarly to several other species. Young adult women and men had similar cell numbers, as opposed to rodents reported to have more KP neurons in the RP3V of females. Human KP neurons differed from the homologous rodent cells as well, in that they were devoid of enkephalins, galanin and tyrosine hydroxylase. Further, they did not contain known KP neuron markers of the human infundibular nucleus, neurokinin B, substance P and cocaine- and amphetamine-regulated transcript, while they received afferent input from these KP neurons. CONCLUSIONS: The identification and positive estrogenic regulation of KP neurons in the human rostral hypothalamus challenge the long-held view that positive estrogen feedback may be restricted to the mediobasal part of the hypothalamus in primates and point to the need of further anatomical, molecular and functional studies of rostral hypothalamic KP neurons.


Asunto(s)
Estrógenos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Menopausia/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Área Preóptica/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Autopsia , Femenino , Humanos , Inmunohistoquímica , Masculino , Microscopía Confocal , Persona de Mediana Edad , Núcleo Hipotalámico Paraventricular/citología , Área Preóptica/citología , Adulto Joven
8.
Dev Comp Immunol ; 90: 41-46, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30179632

RESUMEN

Lumbricin and its orthologue antimicrobial peptides were typically isolated from annelids. In this report, mRNA for lumbricin and -serendipitously- a novel lumbricin-related mRNA sequence were identified in Eisenia andrei earthworms. The determined mRNA sequences of E. andrei lumbricin and lumbricin-related peptide consist of 477 and 575 nucleotides. The precursors of proline-rich E. andrei lumbricin and the related peptide contain 63 and 59 amino acids, respectively. Phylogenetic analysis indicated close relationship with other annelid lumbricins. Highest expression of both mRNAs appeared in the proximal part of the intestine (pharynx, gizzard), while other tested organs had moderate (body wall, midgut, ovary, metanephridium, seminal vesicles, ventral nerve cord) or low (coelomocytes) levels. During ontogenesis their expression revealed continuous increase in embryos. Following 48 h of in vivo Gram-positive bacteria challenge both mRNAs were significantly elevated in coelomocytes, while Gram-negative bacteria or zymosan stimulation had no detectable effects.


Asunto(s)
Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Proteínas del Helminto/genética , Intestinos/fisiología , Oligoquetos/inmunología , Péptidos/genética , Animales , Antiinfecciosos/metabolismo , Clonación Molecular , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas del Helminto/metabolismo , Inmunidad Innata , Oligoquetos/genética , Oligoquetos/microbiología , Péptidos/metabolismo , Filogenia , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA