Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nat Neurosci ; 27(9): 1758-1773, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39095587

RESUMEN

Hypothalamic gonadotropin-releasing hormone (GnRH) neurons regulate fertility and integrate hormonal status with environmental cues to ensure reproductive success. Here we show that GnRH neurons in the olfactory bulb (GnRHOB) of adult mice can mediate social recognition. Specifically, we show that GnRHOB neurons extend neurites into the vomeronasal organ and olfactory epithelium and project to the median eminence. GnRHOB neurons in males express vomeronasal and olfactory receptors, are activated by female odors and mediate gonadotropin release in response to female urine. Male preference for female odors required the presence and activation of GnRHOB neurons, was impaired after genetic inhibition or ablation of these cells and relied on GnRH signaling in the posterodorsal medial amygdala. GnRH receptor expression in amygdala kisspeptin neurons appear to be required for GnRHOB neurons' actions on male mounting behavior. Taken together, these results establish GnRHOB neurons as regulating fertility, sex recognition and mating in male mice.


Asunto(s)
Hormona Liberadora de Gonadotropina , Neuronas , Odorantes , Bulbo Olfatorio , Conducta Sexual Animal , Órgano Vomeronasal , Animales , Masculino , Hormona Liberadora de Gonadotropina/metabolismo , Bulbo Olfatorio/fisiología , Bulbo Olfatorio/metabolismo , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Conducta Sexual Animal/fisiología , Femenino , Órgano Vomeronasal/fisiología , Órgano Vomeronasal/metabolismo , Ratones Endogámicos C57BL , Olfato/fisiología , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología
2.
Front Oncol ; 13: 1240865, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37583932

RESUMEN

Breast carcinoma is the most prevalent cancer among women globally. It has variable clinical courses depending on the stage and clinical-biological features. This case report describes a 56-year-old female with invasive breast cancer without estrogen or progesterone receptor expression, with apocrine differentiation, and with no germline variants in the BRCA1 and BRCA2 genes. Throughout the clinical course, the patient exhibited discordant results for HER2 in immunohistochemistry and in situ hybridization. During the second relapse, the disease displayed apocrine microscopic features. The tumor underwent analysis for the androgen receptor, GCDFP-15, RNA-seq, and whole-genome sequencing (WGS) to identify the breast cancer subtype and to characterize the cancer genome. Our bioinformatic analysis revealed 20,323 somatic SNV/Indels, including five mutations in cancer-related genes that are believed to be responsible for the tumor's development. Two of these mutations were found in the PIK3CA and TP53 genes. Furthermore, the tumor tissue exhibited large copy number alterations to the chromosomes, which could impact gene expression through complex mechanisms and contribute to the tumor phenotype. Clustering algorithms applied on RNA-sequencing data categorized this cancer as a HER2+ subtype. The second-line capecitabine chemotherapy treatment is ongoing, and the patient is responding well. Bioinformatic results support the current treatment decision and open the way to further treatments.

3.
Science ; 377(6610): eabq4515, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36048943

RESUMEN

At the present time, no viable treatment exists for cognitive and olfactory deficits in Down syndrome (DS). We show in a DS model (Ts65Dn mice) that these progressive nonreproductive neurological symptoms closely parallel a postpubertal decrease in hypothalamic as well as extrahypothalamic expression of a master molecule that controls reproduction-gonadotropin-releasing hormone (GnRH)-and appear related to an imbalance in a microRNA-gene network known to regulate GnRH neuron maturation together with altered hippocampal synaptic transmission. Epigenetic, cellular, chemogenetic, and pharmacological interventions that restore physiological GnRH levels abolish olfactory and cognitive defects in Ts65Dn mice, whereas pulsatile GnRH therapy improves cognition and brain connectivity in adult DS patients. GnRH thus plays a crucial role in olfaction and cognition, and pulsatile GnRH therapy holds promise to improve cognitive deficits in DS.


Asunto(s)
Cognición , Disfunción Cognitiva , Síndrome de Down , Hormona Liberadora de Gonadotropina , Trastornos del Olfato , Adulto , Animales , Cognición/efectos de los fármacos , Cognición/fisiología , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Síndrome de Down/complicaciones , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/psicología , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Hormona Liberadora de Gonadotropina/fisiología , Hormona Liberadora de Gonadotropina/uso terapéutico , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Trastornos del Olfato/tratamiento farmacológico , Trastornos del Olfato/etiología , Transmisión Sináptica/efectos de los fármacos , Adulto Joven
4.
Proc Natl Acad Sci U S A ; 119(30): e2203503119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35867816

RESUMEN

Women with polycystic ovary syndrome (PCOS) frequently experience decreased sexual arousal, desire, and sexual satisfaction. While the hypothalamus is known to regulate sexual behavior, the specific neuronal pathways affected in patients with PCOS are not known. To dissect the underlying neural circuitry, we capitalized on a robust preclinical animal model that reliably recapitulates all cardinal PCOS features. We discovered that female mice prenatally treated with anti-Müllerian hormone (PAMH) display impaired sexual behavior and sexual partner preference over the reproductive age. Blunted female sexual behavior was associated with increased sexual rejection and independent of sex steroid hormone status. Structurally, sexual dysfunction was associated with a substantial loss of neuronal nitric oxide synthase (nNOS)-expressing neurons in the ventromedial nucleus of the hypothalamus (VMH) and other areas of hypothalamic nuclei involved in social behaviors. Using in vivo chemogenetic manipulation, we show that nNOSVMH neurons are required for the display of normal sexual behavior in female mice and that pharmacological replenishment of nitric oxide restores normal sexual performance in PAMH mice. Our data provide a framework to investigate facets of hypothalamic nNOS neuron biology with implications for sexual disturbances in PCOS.


Asunto(s)
Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico , Síndrome del Ovario Poliquístico , Conducta Sexual , Núcleo Hipotalámico Ventromedial , Animales , Hormona Antimülleriana/farmacología , Modelos Animales de Enfermedad , Femenino , Preferencia en el Apareamiento Animal , Ratones , Neuronas/efectos de los fármacos , Neuronas/enzimología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Síndrome del Ovario Poliquístico/enzimología , Síndrome del Ovario Poliquístico/fisiopatología , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/metabolismo
5.
EMBO J ; 39(19): e104633, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32761635

RESUMEN

Hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH), the "master molecule" regulating reproduction and fertility, migrate from their birthplace in the nose to their destination using a system of guidance cues, which include the semaphorins and their receptors, the neuropilins and plexins, among others. Here, we show that selectively deleting neuropilin-1 in new GnRH neurons enhances their survival and migration, resulting in excess neurons in the hypothalamus and in their unusual accumulation in the accessory olfactory bulb, as well as an acceleration of mature patterns of activity. In female mice, these alterations result in early prepubertal weight gain, premature attraction to male odors, and precocious puberty. Our findings suggest that rather than being influenced by peripheral energy state, GnRH neurons themselves, through neuropilin-semaphorin signaling, might engineer the timing of puberty by regulating peripheral adiposity and behavioral switches, thus acting as a bridge between the reproductive and metabolic axes.


Asunto(s)
Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/metabolismo , Neuropilina-1/biosíntesis , Conducta Sexual Animal , Maduración Sexual , Aumento de Peso , Animales , Femenino , Hormona Liberadora de Gonadotropina/genética , Masculino , Ratones , Ratones Transgénicos , Neuropilina-1/genética
6.
Dis Model Mech ; 13(3)2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31996360

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons provide a fundamental signal for the onset of puberty and subsequent reproductive functions by secretion of gonadotropin-releasing hormone. Their disrupted development or function leads to congenital hypogonadotropic hypogonadism (CHH). To model the development of human GnRH neurons, we generated a stable GNRH1-TdTomato reporter cell line in human pluripotent stem cells (hPSCs) using CRISPR-Cas9 genome editing. RNA-sequencing of the reporter clone, differentiated into GnRH neurons by dual SMAD inhibition and FGF8 treatment, revealed 6461 differentially expressed genes between progenitors and GnRH neurons. Expression of the transcription factor ISL1, one of the top 50 most upregulated genes in the TdTomato-expressing GnRH neurons, was confirmed in 10.5 gestational week-old human fetal GnRH neurons. Among the differentially expressed genes, we detected 15 genes that are implicated in CHH and several genes that are implicated in human puberty timing. Finally, FGF8 treatment in the neuronal progenitor pool led to upregulation of 37 genes expressed both in progenitors and in TdTomato-expressing GnRH neurons, which suggests upstream regulation of these genes by FGF8 signaling during GnRH neuron differentiation. These results illustrate how hPSC-derived human GnRH neuron transcriptomic analysis can be utilized to dissect signaling pathways and gene regulatory networks involved in human GnRH neuron development.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Genes Reporteros , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Transcriptoma/genética , Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular , Feto/citología , Factor 8 de Crecimiento de Fibroblastos/farmacología , Humanos , Hipogonadismo/genética , Proteínas con Homeodominio LIM/metabolismo , Neuronas/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
7.
Elife ; 82019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31291191

RESUMEN

Congenital hypogonadotropic hypogonadism (CHH) is a condition characterized by absent puberty and infertility due to gonadotropin releasing hormone (GnRH) deficiency, which is often associated with anosmia (Kallmann syndrome, KS). We identified loss-of-function heterozygous mutations in anti-Müllerian hormone (AMH) and its receptor, AMHR2, in 3% of CHH probands using whole-exome sequencing. We showed that during embryonic development, AMH is expressed in migratory GnRH neurons in both mouse and human fetuses and unconvered a novel function of AMH as a pro-motility factor for GnRH neurons. Pathohistological analysis of Amhr2-deficient mice showed abnormal development of the peripheral olfactory system and defective embryonic migration of the neuroendocrine GnRH cells to the basal forebrain, which results in reduced fertility in adults. Our findings highlight a novel role for AMH in the development and function of GnRH neurons and indicate that AMH signaling insufficiency contributes to the pathogenesis of CHH in humans.


Asunto(s)
Hormona Antimülleriana/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipogonadismo/metabolismo , Neuronas/metabolismo , Transducción de Señal , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Hormona Antimülleriana/genética , Axones/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Células COS , Movimiento Celular , Chlorocebus aethiops , Femenino , Fertilidad , Feto/metabolismo , Heterocigoto , Humanos , Mutación con Pérdida de Función , Hormona Luteinizante/metabolismo , Masculino , Ratones Endogámicos C57BL , Bulbo Olfatorio/metabolismo , Linaje , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Adulto Joven
8.
Brain Res ; 1688: 8-14, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29555237

RESUMEN

The kisspeptin system is clustered in two main groups of cell bodies (the periventricular region, RP3V and the arcuate nucleus, ARC) that send fibers mainly to the GnRH neurons and in a few other locations, including the paraventricular nucleus, PVN. In physiological conditions, gonadal hormones modulate the kisspeptin system with expression changes according to different phases of the estrous cycle: the highest being in estrus phase in RP3V and PVN (positive feedback), and in ARC during the diestrus phase (negative feedback). In this work we wanted to study these hormonal fluctuations during the estrous cycle, investigating the role played by progesterone (P) or estradiol (E2), alone or together, on the kisspeptin system. Gonadectomized CD1 female mice were treated with P, E2 or both (E2 + P), following a timing of administration that emulates the different phases of estrous cycle, for two cycles of 4 days. As expected, the two cell groups were differentially affected by E2; the RP3V group was positively influenced by E2 (alone or with the P), whereas in the ARC the administration of E2 did not affect the system. However P (alone) induced a rise in the kisspeptin immunoreactivity. All the treatments significantly affected the kisspeptin innervation of the PVN, with regional differences, suggesting that these fibers arrive from both RP3V and ARC nuclei.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Estradiol/metabolismo , Ciclo Estral , Kisspeptinas/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Progesterona/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Estradiol/administración & dosificación , Femenino , Ratones , Ovariectomía , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Progesterona/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA