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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 128(10): 1941-1954, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36959380

RESUMEN

BACKGROUND: Systemic therapy for metastatic clear cell sarcoma (CCS) bearing EWSR1-CREB1/ATF1 fusions remains an unmet clinical need in children, adolescents, and young adults. METHODS: To identify key signaling pathway vulnerabilities in CCS, a multi-pronged approach was taken: (i) genomic and transcriptomic landscape analysis, (ii) integrated chemical biology interrogations, (iii) development of CREB1/ATF1 inhibitors, and (iv) antibody-drug conjugate testing (ADC). The first approach encompassed DNA exome and RNA deep sequencing of the largest human CCS cohort yet reported consisting of 47 patient tumor samples and 8 cell lines. RESULTS: Sequencing revealed recurrent mutations in cell cycle checkpoint, DNA double-strand break repair or DNA mismatch repair genes, with a correspondingly low to intermediate tumor mutational burden. DNA multi-copy gains with corresponding high RNA expression were observed in CCS tumor subsets. CCS cell lines responded to the HER3 ADC patritumab deruxtecan in a dose-dependent manner in vitro, with impaired long term cell viability. CONCLUSION: These studies of the genomic, transcriptomic and chemical biology landscape represent a resource 'atlas' for the field of CCS investigation and drug development. CHK inhibitors are identified as having potential relevance, CREB1 inhibitors non-dependence of CCS on CREB1 activity was established, and the potential utility of HER3 ADC being used in CCS is found.


Asunto(s)
Sarcoma de Células Claras , Niño , Adolescente , Adulto Joven , Humanos , Sarcoma de Células Claras/genética , Sarcoma de Células Claras/metabolismo , Sarcoma de Células Claras/patología , Transcriptoma , Genómica , Secuencia de Bases , ARN , Proteínas de Fusión Oncogénica/genética
2.
BMC Genomics ; 18(1): 411, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28545403

RESUMEN

BACKGROUND: Reduced physical activity and increased intake of calorically-dense diets are the main risk factors for obesity, glucose intolerance, and type 2 diabetes. Chronic overnutrition and hyperglycemia can alter gene expression, contributing to long-term obesity complications. While caloric restriction can reduce obesity and glucose intolerance, it is currently unknown whether it can effectively reprogram transcriptome to a pre-obesity level. The present study addressed this question by the preliminary examination of the transcriptional dynamics in skeletal muscle after exposure to overnutrition and following caloric restriction. RESULTS: Six male rhesus macaques of 12-13 years of age consumed a high-fat western-style diet for 6 months and then were calorically restricted for 4 months without exercise. Skeletal muscle biopsies were subjected to longitudinal gene expression analysis using next-generation whole-genome RNA sequencing. In spite of significant weight loss and normalized insulin sensitivity, the majority of WSD-induced (n = 457) and WSD-suppressed (n = 47) genes remained significantly dysregulated after caloric restriction (FDR ≤0.05). The MetacoreTM pathway analysis reveals that western-style diet induced the sustained activation of the transforming growth factor-ß gene network, associated with extracellular matrix remodeling, and the downregulation of genes involved in muscle structure development and nutritional processes. CONCLUSIONS: Western-style diet, in the absence of exercise, induced skeletal muscle transcriptional programing, which persisted even after insulin resistance and glucose intolerance were completely reversed with caloric restriction.


Asunto(s)
Perfilación de la Expresión Génica , Músculo Esquelético/metabolismo , Obesidad/genética , Animales , Restricción Calórica , Citocinas/sangre , Dieta Occidental/efectos adversos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Macaca mulatta , Masculino , Músculo Esquelético/efectos de los fármacos , Obesidad/inducido químicamente , Obesidad/metabolismo , Obesidad/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/efectos de los fármacos
3.
Front Neuroendocrinol ; 36: 90-107, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25171849

RESUMEN

Substantial progress has been made in recent years toward deciphering the molecular and genetic underpinnings of the pubertal process. The availability of powerful new methods to interrogate the human genome has led to the identification of genes that are essential for puberty to occur. Evidence has also emerged suggesting that the initiation of puberty requires the coordinated activity of gene sets organized into functional networks. At a cellular level, it is currently thought that loss of transsynaptic inhibition, accompanied by an increase in excitatory inputs, results in the pubertal activation of GnRH release. This concept notwithstanding, a mechanism of epigenetic repression targeting genes required for the pubertal activation of GnRH neurons was recently identified as a core component of the molecular machinery underlying the central restraint of puberty. In this chapter we will discuss the potential contribution of various mechanisms of epigenetic regulation to the hypothalamic control of female puberty.


Asunto(s)
Epigénesis Genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/fisiología , Neuronas/metabolismo , Pubertad/fisiología , Maduración Sexual/fisiología , Animales , Femenino , Humanos
4.
Neuroendocrinology ; 99(2): 94-107, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24686008

RESUMEN

The importance of the Kiss1 gene in the control of reproductive development is well documented. However, much less is known about the transcriptional regulation of Kiss1 expression in the hypothalamus. Critical for these studies is an accurate identification of the site(s) where Kiss1 transcription is initiated. Employing 5'-RACE PCR, we detected a transcription start site (TSS1) used by the hypothalamus of rats, mice, nonhuman primates and humans to initiate Kiss1 transcription. In rodents, an exon 1 encoding 5'-untranslated sequences is followed by an alternatively spliced second exon, which encodes 5'-untranslated regions of two different lengths and contains the translation initiation codon (ATG). In nonhuman primates and humans, exon 2 is not alternatively spliced. Surprisingly, in rat mediobasal hypothalamus (MBH), but not preoptic area (POA), an additional TSS (TSS2) located upstream from TSS1 generates an exon 1 longer (377 bp) than the TSS1-derived exon 1 (98 bp). The content of TSS1-derived transcripts increased at puberty in the POA and MBH of female rats. It also increased in the MBH after ovariectomy, and this change was prevented by estrogen. In contrast, no such changes in TSS2-derived transcript abundance were detected. Promoter assays showed that the proximal TSS1 promoter is much more active than the putative TSS2 promoter, and that only the TSS1 promoter is regulated by estrogen. These differences appear to be related to the presence of a TATA box and binding sites for transcription factors activating transcription and interacting with estrogen receptor-α in the TSS1, but not TSS2, promoter.


Asunto(s)
Estrógenos/farmacología , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , ARN Mensajero/metabolismo , Maduración Sexual , Sitio de Iniciación de la Transcripción , Transcripción Genética/efectos de los fármacos , Animales , Receptor alfa de Estrógeno/efectos de los fármacos , Terapia de Reemplazo de Estrógeno , Exones/genética , Femenino , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Ovariectomía , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley , Transcripción Genética/genética
5.
Horm Behav ; 64(2): 175-86, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23998662

RESUMEN

This article is part of a Special Issue "Puberty and Adolescence". Puberty is a major developmental milestone controlled by the interaction of genetic factors and environmental cues of mostly metabolic and circadian nature. An increased pulsatile release of the decapeptide gonadotropin releasing hormone (GnRH) from hypothalamic neurosecretory neurons is required for both the initiation and progression of the pubertal process. This increase is brought about by coordinated changes that occur in neuronal and glial networks associated with GnRH neurons. These changes ultimately result in increased neuronal and glial stimulatory inputs to the GnRH neuronal network and a reduction of transsynaptic inhibitory influences. While some of the major players controlling pubertal GnRH secretion have been identified using gene-centric approaches, much less is known about the system-wide control of the overall process. Because the pubertal activation of GnRH release involves a diversity of cellular phenotypes, and a myriad of intracellular and cell-to-cell signaling molecules, it appears that the overall process is controlled by a highly coordinated and interactive regulatory system involving hundreds, if not thousands, of gene products. In this article we will discuss emerging evidence suggesting that these genes are arranged as functionally connected networks organized, both internally and across sub-networks, in a hierarchical fashion. According to this concept, the core of these networks is composed of transcriptional regulators that, by directing expression of downstream subordinate genes, provide both stability and coordination to the cellular networks involved in initiating the pubertal process. The integrative response of these gene networks to external inputs is postulated to be coordinated by epigenetic mechanisms.


Asunto(s)
Redes Reguladoras de Genes , Sistemas Neurosecretores/fisiología , Primates/fisiología , Maduración Sexual/genética , Biología de Sistemas/métodos , Animales , Epigénesis Genética/fisiología , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratas
7.
Clin Transl Med ; 12(7): e961, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35839307

RESUMEN

BACKGROUND: Metastatic epithelioid sarcoma (EPS) remains a largely unmet clinical need in children, adolescents and young adults despite the advent of EZH2 inhibitor tazemetostat. METHODS: In order to realise consistently effective drug therapies, a functional genomics approach was used to identify key signalling pathway vulnerabilities in a spectrum of EPS patient samples. EPS biopsies/surgical resections and cell lines were studied by next-generation DNA exome and RNA deep sequencing, then EPS cell cultures were tested against a panel of chemical probes to discover signalling pathway targets with the most significant contributions to EPS tumour cell maintenance. RESULTS: Other biologically inspired functional interrogations of EPS cultures using gene knockdown or chemical probes demonstrated only limited to modest efficacy in vitro. However, our molecular studies uncovered distinguishing features (including retained dysfunctional SMARCB1 expression and elevated GLI3, FYN and CXCL12 expression) of distal, paediatric/young adult-associated EPS versus proximal, adult-associated EPS. CONCLUSIONS: Overall results highlight the complexity of the disease and a limited chemical space for therapeutic advancement. However, subtle differences between the two EPS subtypes highlight the biological disparities between younger and older EPS patients and emphasise the need to approach the two subtypes as molecularly and clinically distinct diseases.


Asunto(s)
Proteínas de Unión al ADN , Sarcoma , Adolescente , Niño , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/uso terapéutico , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/uso terapéutico , Genómica , Humanos , Sarcoma/tratamiento farmacológico , Sarcoma/genética , Sarcoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/uso terapéutico , Adulto Joven
8.
Sci Rep ; 11(1): 1996, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479437

RESUMEN

Female puberty is subject to Polycomb Group (PcG)-dependent transcriptional repression. Kiss1, a puberty-activating gene, is a key target of this silencing mechanism. Using a gain-of-function approach and a systems biology strategy we now show that EED, an essential PcG component, acts in the arcuate nucleus of the hypothalamus to alter the functional organization of a gene network involved in the stimulatory control of puberty. A central node of this network is Kdm6b, which encodes an enzyme that erases the PcG-dependent histone modification H3K27me3. Kiss1 is a first neighbor in the network; genes encoding glutamatergic receptors and potassium channels are second neighbors. By repressing Kdm6b expression, EED increases H3K27me3 abundance at these gene promoters, reducing gene expression throughout a gene network controlling puberty activation. These results indicate that Kdm6b repression is a basic mechanism used by PcG to modulate the biological output of puberty-activating gene networks.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/genética , Kisspeptinas/genética , Complejo Represivo Polycomb 2/genética , Pubertad/genética , Animales , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Neuronas/metabolismo , Sistemas Neurosecretores/crecimiento & desarrollo , Sistemas Neurosecretores/metabolismo , Proteínas del Grupo Polycomb/genética , Regiones Promotoras Genéticas/genética , Pubertad/fisiología , Ratas , Biología de Sistemas
9.
Nutrients ; 12(2)2020 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-32085416

RESUMEN

White adipose tissue (WAT) hypertrophy is an essential hallmark of obesity and is associated with the activation of resident immune cells. While the benefits of caloric restriction (CR) on health span are generally accepted, its effects on WAT physiology are not well understood. We previously demonstrated that short-term CR reverses obesity in male rhesus macaques exposed to a high-fat Western-style diet (WSD). Here, we analyzed subcutaneous WAT biopsies collected from this cohort of animals before and after WSD and following CR. This analysis showed that WSD induced adipocyte hypertrophy and inhibited ß-adrenergic-simulated lipolysis. CR reversed adipocyte hypertrophy, but WAT remained insensitive to ß-adrenergic agonist stimulation. Whole-genome transcriptional analysis revealed that ß3-adrenergic receptor and de novo lipogenesis genes were downregulated by WSD and remained downregulated after CR. In contrast, WSD-induced pro-inflammatory gene expression was effectively reversed by CR. Furthermore, peripheral blood monocytes isolated during the CR period exhibited a significant reduction in the production of pro-inflammatory cytokines compared to those obtained after WSD. Collectively, this study demonstrates that short-term CR eliminates an obesity-induced pro-inflammatory response in WAT and peripheral monocytes.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Restricción Calórica , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Mediadores de Inflamación/metabolismo , Obesidad/dietoterapia , Obesidad/metabolismo , Tejido Adiposo Blanco/patología , Agonistas Adrenérgicos beta , Animales , Regulación hacia Abajo , Hipertrofia/dietoterapia , Hipertrofia/etiología , Lipogénesis/genética , Lipólisis , Macaca mulatta , Masculino , Monocitos/metabolismo , Obesidad/etiología , Obesidad/patología , Receptores Adrenérgicos beta 3/genética
10.
PLoS One ; 15(4): e0231826, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32324802

RESUMEN

The affordability of next-generation genomic sequencing and the improvement of medical data management have contributed largely to the evolution of biological analysis from both a clinical and research perspective. Precision medicine is a response to these advancements that places individuals into better-defined subsets based on shared clinical and genetic features. The identification of personalized diagnosis and treatment options is dependent on the ability to draw insights from large-scale, multi-modal analysis of biomedical datasets. Driven by a real use case, we premise that platforms that support precision medicine analysis should maintain data in their optimal data stores, should support distributed storage and query mechanisms, and should scale as more samples are added to the system. We extended a genomics-based columnar data store, GenomicsDB, for ease of use within a distributed analytics platform for clinical and genomic data integration, known as the ODA framework. The framework supports interaction from an i2b2 plugin as well as a notebook environment. We show that the ODA framework exhibits worst-case linear scaling for array size (storage), import time (data construction), and query time for an increasing number of samples. We go on to show worst-case linear time for both import of clinical data and aggregate query execution time within a distributed environment. This work highlights the integration of a distributed genomic database with a distributed compute environment to support scalable and efficient precision medicine queries from a HIPAA-compliant, cohort system in a real-world setting. The ODA framework is currently deployed in production to support precision medicine exploration and analysis from clinicians and researchers at UCLA David Geffen School of Medicine.


Asunto(s)
Genómica , Selección de Paciente , Medicina de Precisión/métodos , Estudios de Cohortes , Bases de Datos Genéticas , Conjuntos de Datos como Asunto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
11.
Nat Commun ; 9(1): 57, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29302059

RESUMEN

Polycomb group (PcG) proteins control the timing of puberty by repressing the Kiss1 gene in hypothalamic arcuate nucleus (ARC) neurons. Here we identify two members of the Trithorax group (TrxG) of modifiers, mixed-lineage leukemia 1 (MLL1), and 3 (MLL3), as central components of an activating epigenetic machinery that dynamically counteracts PcG repression. Preceding puberty, MLL1 changes the chromatin configuration at the promoters of Kiss1 and Tac3, two genes required for puberty to occur, from repressive to permissive. Concomitantly, MLL3 institutes a chromatin structure that changes the functional status of a Kiss1 enhancer from poised to active. RNAi-mediated, ARC-specific Mll1 knockdown reduced Kiss1 and Tac3 expression, whereas CRISPR-Cas9-directed epigenome silencing of the Kiss1 enhancer selectively reduced Kiss1 activity. Both interventions delay puberty and disrupt reproductive cyclicity. Our results demonstrate that an epigenetic switch from transcriptional repression to activation is crucial to the regulatory mechanism controlling the timing of mammalian puberty.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Hipotálamo/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Pubertad/genética , Animales , Sistemas CRISPR-Cas , Cromatina , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Kisspeptinas/genética , Macaca mulatta , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Taquicininas/genética
12.
Nat Commun ; 9(1): 1977, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773799

RESUMEN

Population studies elucidating the genetic architecture of reproductive ageing have been largely limited to European ancestries, restricting the generalizability of the findings and overlooking possible key genes poorly captured by common European genetic variation. Here, we report 26 loci (all P < 5 × 10-8) for reproductive ageing, i.e. puberty timing or age at menopause, in a non-European population (up to 67,029 women of Japanese ancestry). Highlighted genes for menopause include GNRH1, which supports a primary, rather than passive, role for hypothalamic-pituitary GnRH signalling in the timing of menopause. For puberty timing, we demonstrate an aetiological role for receptor-like protein tyrosine phosphatases by combining evidence across population genetics and pre- and peri-pubertal changes in hypothalamic gene expression in rodent and primate models. Furthermore, our findings demonstrate widespread differences in allele frequencies and effect estimates between Japanese and European associated variants, highlighting the benefits and challenges of large-scale trans-ethnic approaches.


Asunto(s)
Envejecimiento/genética , Pueblo Asiatico/genética , Sitios Genéticos/fisiología , Menarquia/genética , Menopausia/genética , Adolescente , Adulto , Factores de Edad , Animales , Niño , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Frecuencia de los Genes/fisiología , Variación Genética/fisiología , Humanos , Hipotálamo/metabolismo , Japón , Macaca mulatta , Metaanálisis como Asunto , Persona de Mediana Edad , Modelos Animales , Ratas Sprague-Dawley , Población Blanca/genética
13.
Endocrinology ; 148(11): 5147-61, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17615149

RESUMEN

Much has been learned in recent years about the central mechanisms controlling the initiation of mammalian puberty. It is now clear that this process requires the interactive participation of several genes. Using a combination of high throughput, molecular, and bioinformatics strategies, in combination with a system biology approach, we singled out from the hypothalamus of nonhuman primates and rats a group of related genes whose expression increases at the time of female puberty. Although these genes [henceforth termed tumor-related genes (TRGs)] have diverse cellular functions, they share the common feature of having been earlier identified as involved in tumor suppression/tumor formation. A prominent member of this group is KiSS1, a gene recently shown to be essential for the occurrence of puberty. Cis-regulatory analysis revealed the presence of a hierarchically arranged gene set containing five major hubs (CDP/CUTL1, MAF, p53, YY1, and USF2) controlling the network at the transcriptional level. In turn, these hubs are heavily connected to non-TRGs involved in the transcriptional regulation of the pubertal process. TRGs may be expressed in the mammalian hypothalamus as components of a regulatory gene network that facilitates and integrates cellular and cell-cell communication programs required for the acquisition of female reproductive competence.


Asunto(s)
Redes Reguladoras de Genes , Genes Relacionados con las Neoplasias , Hipotálamo/metabolismo , Maduración Sexual/genética , Regulación hacia Arriba , Región de Flanqueo 5' , Animales , Sitios de Unión , Femenino , Regulación del Desarrollo de la Expresión Génica , Macaca mulatta , Modelos Biológicos , Sistemas Neurosecretores/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/metabolismo
14.
BMC Genomics ; 8: 43, 2007 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-17286860

RESUMEN

BACKGROUND: Rhesus macaques serve a critical role in the study of human biomedical research. While both Indian and Chinese rhesus macaques are commonly used, genetic differences between these two subspecies affect aspects of their behavior and physiology, including response to simian immunodeficiency virus (SIV) infection. Single nucleotide polymorphisms (SNPs) can play an important role in both establishing ancestry and in identifying genes involved in complex diseases. We sequenced the 3' end of rhesus macaque genes in an effort to identify gene-based SNPs that could distinguish between Indian and Chinese rhesus macaques and aid in association analysis. RESULTS: We surveyed the 3' end of 94 genes in 20 rhesus macaque animals. The study included 10 animals each of Indian and Chinese ancestry. We identified a total of 661 SNPs, 457 of which appeared exclusively in one or the other population. Seventy-nine additional animals were genotyped at 44 of the population-exclusive SNPs. Of those, 38 SNPs were confirmed as being population-specific. CONCLUSION: This study demonstrates that the 3' end of genes is rich in sequence polymorphisms and is suitable for the efficient discovery of gene-linked SNPs. In addition, the results show that the genomic sequences of Indian and Chinese rhesus macaque are remarkably divergent, and include numerous population-specific SNPs. These ancestral SNPs could be used for the rapid scanning of rhesus macaques, both to establish animal ancestry and to identify gene alleles that may contribute to the phenotypic differences observed in these populations.


Asunto(s)
Macaca mulatta/genética , Polimorfismo de Nucleótido Simple , Animales , China , Genética de Población , Genotipo , India , Modelos Biológicos , Especificidad de la Especie
15.
Endocrinology ; 157(8): 3224-32, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27355492

RESUMEN

Llamas are considered to be reflex ovulators. However, semen from these animals is reported to be rich in ovulation-inducing factor(s), one of which has been identified as nerve growth factor (NGF). These findings suggest that ovulation in llamas may be elicited by chemical signals contained in semen instead of being mediated by neural signals. The present study examines this notion. Llamas displaying a preovulatory follicle were assigned to four groups: group 1 received an intrauterine infusion (IUI) of PBS; group 2 received an IUI of seminal plasma; group 3 was mated to a male whose urethra had been surgically diverted (urethrostomized male); and group 4 was mated to an intact male. Ovulation (detected by ultrasonography) occurred only in llamas mated to an intact male or given an IUI of seminal plasma and was preceded by a surge in plasma LH levels initiated within an hour after coitus or IUI. In both ovulatory groups, circulating ß-NGF levels increased within 15 minutes after treatment, reaching values that were greater and more sustained in llamas mated with an intact male. These results demonstrate that llamas can be induced to ovulate by seminal plasma in the absence of copulation and that copulation alone cannot elicit ovulation in the absence of seminal plasma. In addition, our results implicate ß-NGF as an important mediator of seminal plasma-induced ovulation in llamas because ovulation does not occur if ß-NGF levels do not increase in the bloodstream, a change that occurs promptly after copulation with an intact male or IUI of seminal plasma.


Asunto(s)
Camélidos del Nuevo Mundo/fisiología , Factor de Crecimiento Nervioso/farmacología , Inducción de la Ovulación/veterinaria , Semen/fisiología , Animales , Copulación , Cuerpo Lúteo/diagnóstico por imagen , Cuerpo Lúteo/fisiología , Femenino , Inseminación Artificial/métodos , Inseminación Artificial/veterinaria , Hormona Luteinizante/sangre , Masculino , Factor de Crecimiento Nervioso/sangre , Folículo Ovárico/citología , Folículo Ovárico/diagnóstico por imagen , Ovulación/efectos de los fármacos , Ovulación/fisiología , Progesterona/sangre , Ultrasonografía
16.
Nat Commun ; 6: 10195, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26671628

RESUMEN

In primates, puberty is unleashed by increased GnRH release from the hypothalamus following an interval of juvenile quiescence. GWAS implicates Zinc finger (ZNF) genes in timing human puberty. Here we show that hypothalamic expression of several ZNFs decreased in agonadal male monkeys in association with the pubertal reactivation of gonadotropin secretion. Expression of two of these ZNFs, GATAD1 and ZNF573, also decreases in peripubertal female monkeys. However, only GATAD1 abundance increases when gonadotropin secretion is suppressed during late infancy. Targeted delivery of GATAD1 or ZNF573 to the rat hypothalamus delays puberty by impairing the transition of a transcriptional network from an immature repressive epigenetic configuration to one of activation. GATAD1 represses transcription of two key puberty-related genes, KISS1 and TAC3, directly, and reduces the activating histone mark H3K4me2 at each promoter via recruitment of histone demethylase KDM1A. We conclude that GATAD1 epitomizes a subset of ZNFs involved in epigenetic repression of primate puberty.


Asunto(s)
Epigénesis Genética , Factores de Transcripción GATA/genética , Regulación del Desarrollo de la Expresión Génica , Hipotálamo/metabolismo , Pubertad/genética , ARN Mensajero/metabolismo , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Femenino , Técnica del Anticuerpo Fluorescente , Hormona Folículo Estimulante/metabolismo , Factores de Transcripción GATA/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Histona Demetilasas/metabolismo , Hibridación Fluorescente in Situ , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Macaca mulatta , Masculino , Neuroquinina B/genética , Neuroquinina B/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Dedos de Zinc/genética
17.
J Rare Disord ; 1(2): 37-42, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28824928

RESUMEN

BACKGROUND: Osteogenesis imperfecta (OI) type V is a dominantly inherited skeletal dysplasia characterized by fractures and progressive deformity of long bones. In addition, patients often present with radial head dislocation, hyperplastic callus, and calcification of the forearm interosseous membrane. Recently, a specific mutation in the IFITM5 gene was found to be responsible for OI type V. This mutation, a C to T transition 14 nucleotides upstream from the endogenous start codon, creates a new start methionine that appears to be preferentially used by the translational machinery. However, the mechanism by which the lengthened protein results in a dominant type of OI is unknown. METHODS AND RESULTS: We report 7 ethnically diverse (African-American, Caucasian, Hispanic, and African) individuals with OI type V from 2 families and 2 sporadic cases. Exome sequencing failed to identify a causative mutation. Using Sanger sequencing, we found that all affected individuals in our cohort possess the c.-14 IFITM5 variant, further supporting the notion that OI type V is caused by a single, discrete mutation. Our patient cohort demonstrated inter-and intrafamilial phenotypic variability, including a father with classic OI type V whose daughter had a phenotype similar to OI type I. This clinical variability suggests that modifier genes influence the OI type V phenotype. We also confirm that the mutation creates an aberrant IFITM5 protein containing an additional 5 amino acids at the N-terminus. CONCLUSIONS: The variable clinical signs in these cases illustrate the significant variability of the OI type V phenotype caused by the c.-14 IFITM5 mutation. The affected individuals are more ethnically diverse than previously reported.

18.
Nat Neurosci ; 16(3): 281-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23354331

RESUMEN

The timing of puberty is controlled by many genes. The elements coordinating this process have not, however, been identified. Here we show that an epigenetic mechanism of transcriptional repression times the initiation of female puberty in rats. We identify silencers of the Polycomb group (PcG) as principal contributors to this mechanism and show that PcG proteins repress Kiss1, a puberty-activating gene. Hypothalamic expression of two key PcG genes, Eed and Cbx7, decreased and methylation of their promoters increased before puberty. Inhibiting DNA methylation blocked both events and resulted in pubertal failure. The pubertal increase in Kiss1 expression was accompanied by EED loss from the Kiss1 promoter and enrichment of histone H3 modifications associated with gene activation. Preventing the eviction of EED from the Kiss1 promoter disrupted pulsatile gonadotropin-releasing hormone release, delayed puberty and compromised fecundity. Our results identify epigenetic silencing as a mechanism underlying the neuroendocrine control of female puberty.


Asunto(s)
Epigénesis Genética , Hipotálamo/fisiología , Maduración Sexual/fisiología , Animales , Metilación de ADN , Estradiol/sangre , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Histonas/genética , Histonas/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Ratas , Ratas Sprague-Dawley
19.
Horm Res Paediatr ; 80(4): 257-66, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24051510

RESUMEN

BACKGROUND/AIM: TTF1 and EAP1 are transcription factors that modulate gonadotropin-releasing hormone expression. We investigated the contribution of TTF1 and EAP1 genes to central pubertal disorders. PATIENTS AND METHODS: 133 patients with central pubertal disorders were studied: 86 with central precocious puberty and 47 with normosmic isolated hypogonadotropic hypogonadism. The coding region of TTF1 and EAP1 were sequenced. Variations of polyglutamine and polyalanine repeats in EAP1 were analyzed by GeneScan software. Association of TTF1 and EAP1 to genes implicated in timing of puberty was investigated by meta-network framework GeneMANIA and Cytoscape software. RESULTS: Direct sequencing of the TTF1 did not reveal any mutation or polymorphisms. Four EAP1 synonymous variants were identified with similar frequencies among groups. The most common EAP1 5'-distal polyalanine genotype was the homozygous 12/12, but the genotype 12/9 was identified in 2 central precocious puberty sisters without functional alteration in EAP1 transcriptional activity. TTF1 and EAP1 were connected, via genetic networks, to genes implicated in the control of menarche. CONCLUSION: No TTF1 or EAP1 germline mutations were associated with central pubertal disorders. TTF1 and EAP1 may affect puberty by changing expression in response to other members of puberty-associated gene networks, or by differentially affecting the expression of gene components of these networks.


Asunto(s)
Proteínas de Unión al ADN , Regulación de la Expresión Génica/genética , Hormona Liberadora de Gonadotropina , Hipogonadismo , Securina , Niño , Análisis Mutacional de ADN , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Femenino , Genotipo , Mutación de Línea Germinal , Humanos , Hipogonadismo/genética , Hipogonadismo/metabolismo , Masculino , Securina/biosíntesis , Securina/genética , Factores de Transcripción
20.
PLoS One ; 6(11): e26160, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22073148

RESUMEN

BACKGROUND: Computational prediction of Transcription Factor Binding Sites (TFBS) from sequence data alone is difficult and error-prone. Machine learning techniques utilizing additional environmental information about a predicted binding site (such as distances from the site to particular chromatin features) to determine its occupancy/functionality class show promise as methods to achieve more accurate prediction of true TFBS in silico. We evaluate the Bayesian Network (BN) and Support Vector Machine (SVM) machine learning techniques on four distinct TFBS data sets and analyze their performance. We describe the features that are most useful for classification and contrast and compare these feature sets between the factors. RESULTS: Our results demonstrate good performance of classifiers both on TFBS for transcription factors used for initial training and for TFBS for other factors in cross-classification experiments. We find that distances to chromatin modifications (specifically, histone modification islands) as well as distances between such modifications to be effective predictors of TFBS occupancy, though the impact of individual predictors is largely TF specific. In our experiments, Bayesian network classifiers outperform SVM classifiers. CONCLUSIONS: Our results demonstrate good performance of machine learning techniques on the problem of occupancy classification, and demonstrate that effective classification can be achieved using distances to chromatin features. We additionally demonstrate that cross-classification of TFBS is possible, suggesting the possibility of constructing a generalizable occupancy classifier capable of handling TFBS for many different transcription factors.


Asunto(s)
Factores de Transcripción/metabolismo , Algoritmos , Teorema de Bayes , Sitios de Unión , Cromosomas Humanos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA