Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioengineered ; 13(3): 4832-4843, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35156513

RESUMEN

Long non-coding (lnc) RNA serves a vital role in the cellular processes of carcinoma. This study aimed to explore the accurate mechanism underlying lncRNA small nucleolar RNA host gene 8 (SNHG8) in melanoma. In this study, lncRNA SNHG8 expression were upregulated in melanoma tissues and cells, and lncRNA SNHG8 knockdown reduced melanoma cell viability, migration and invasion. Moreover, lncRNA SNHG8 expression could be induced by transcription factor YY1. In addition, we found that miR-656 could directly bind to lncRNA SNHG8 and SERPINE1 mRNA binding protein 1 (SERBP1). Rescue assays indicated that miR-656 overexpression inhibited the aforementioned cellular activities in melanoma cells, which were reversed by SERBP1 overexpression. In conclusion, this work elucidated that YY1-induced upregulation of lncRNA SNHG8 boosted the development of melanoma via the miR-656-3p/SERBP1 axis, providing a novel therapeutic strategy for melanoma treatment.


Asunto(s)
Melanoma , MicroARNs , ARN Largo no Codificante , Proteínas de Unión al ARN , Factor de Transcripción YY1 , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Melanoma/genética , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Mensajero , ARN Nucleolar Pequeño , Proteínas de Unión al ARN/genética , Factor de Transcripción YY1/genética
2.
Int J Mol Sci ; 22(18)2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-34575930

RESUMEN

Low body weight at birth has been shown to be a risk factor for future metabolic disorders, as well as stress response abnormalities and depression. We showed that low-birthweight rats had prolonged high blood corticosterone levels after stress exposure, and that an increase in Gas5 lncRNA, a decoy receptor for glucocorticoid receptors (GRs), reduces glucocorticoid responsiveness. Thus, we concluded that dampened pituitary glucocorticoid responsiveness disturbed the glucocorticoid feedback loop in low-birthweight rats. However, it remains unclear whether such glucocorticoid responsiveness is suppressed solely in the pituitary or systemically. The expression of Gas5 lncRNA increased only in the pituitary, and the intact induction of expression of the GR co-chaperone factor Fkbp5 against dexamethasone was seen in the liver, muscle, and adipose tissue. Intervention with a methyl-modulator diet (folate, VB12, choline, betaine, and zinc) immediately before or one week after delivery reversed the expression level of Gas5 lncRNA in the pituitary of the offspring. Consequently, it partially normalized the blood corticosterone levels after restraint stress exposure. In conclusion, the mode of glucocorticoid response in low-birthweight rats is impaired solely in the pituitary, and intervention with methyl-modulators ameliorates the impairment, but with a narrow therapeutic time window.


Asunto(s)
Biomarcadores , Peso al Nacer , Glucocorticoides/metabolismo , Estrés Fisiológico , Estrés Psicológico , Animales , Suplementos Dietéticos , Femenino , Expresión Génica , Glucocorticoides/sangre , Metilación , Especificidad de Órganos , Hipófisis/metabolismo , Embarazo , ARN Nucleolar Pequeño/genética , Ratas , Receptores de Glucocorticoides/metabolismo
3.
Hum Mol Genet ; 30(12): 1101-1110, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-33856031

RESUMEN

The smallest genomic region causing Prader-Willi Syndrome (PWS) deletes the non-coding RNA SNORD116 cluster; however, the function of SNORD116 remains a mystery. Previous work in the field revealed the tantalizing possibility that expression of NHLH2, a gene previously implicated in both obesity and hypogonadism, was downregulated in PWS patients and differentiated stem cells. In silico RNA: RNA modeling identified several potential interaction domains between SNORD116 and NHLH2 mRNA. One of these interaction domains was highly conserved in most vertebrate NHLH2 mRNAs examined. A construct containing the Nhlh2 mRNA, including its 3'-UTR, linked to a c-myc tag was transfected into a hypothalamic neuron cell line in the presence and absence of exogenously-expressed Snord116. Nhlh2 mRNA expression was upregulated in the presence of Snord116 dependent on the length and type of 3'UTR used on the construct. Furthermore, use of actinomycin D to stop new transcription in N29/2 cells demonstrated that the upregulation occurred through increased stability of the Nhlh2 mRNA in the 45 minutes immediately following transcription. In silico modeling also revealed that a single nucleotide variant (SNV) in the NHLH2 mRNA could reduce the predicted interaction strength of the NHLH2:SNORD116 diad. Indeed, use of an Nhlh2 mRNA construct containing this SNV significantly reduces the ability of Snord116 to increase Nhlh2 mRNA levels. For the first time, these data identify a motif and mechanism for SNORD116-mediated regulation of NHLH2, clarifying the mechanism by which deletion of the SNORD116 snoRNAs locus leads to PWS phenotypes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Síndrome de Prader-Willi/genética , Proteínas Proto-Oncogénicas c-myc/genética , ARN Nucleolar Pequeño/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Ratones , Neuronas/metabolismo , Neuronas/patología , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patología , Procesamiento Postranscripcional del ARN/genética , Estabilidad del ARN/genética
4.
JCI Insight ; 5(12)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32365348

RESUMEN

Imprinted genes are highly expressed in the hypothalamus; however, whether specific imprinted genes affect hypothalamic neuromodulators and their functions is unknown. It has been suggested that Prader-Willi syndrome (PWS), a neurodevelopmental disorder caused by lack of paternal expression at chromosome 15q11-q13, is characterized by hypothalamic insufficiency. Here, we investigate the role of the paternally expressed Snord116 gene within the context of sleep and metabolic abnormalities of PWS, and we report a significant role of this imprinted gene in the function and organization of the 2 main neuromodulatory systems of the lateral hypothalamus (LH) - namely, the orexin (OX) and melanin concentrating hormone (MCH) - systems. We observed that the dynamics between neuronal discharge in the LH and the sleep-wake states of mice with paternal deletion of Snord116 (PWScrm+/p-) are compromised. This abnormal state-dependent neuronal activity is paralleled by a significant reduction in OX neurons in the LH of mutant mice. Therefore, we propose that an imbalance between OX- and MCH-expressing neurons in the LH of mutant mice reflects a series of deficits manifested in the PWS, such as dysregulation of rapid eye movement (REM) sleep, food intake, and temperature control.


Asunto(s)
Conducta Animal/fisiología , Área Hipotalámica Lateral/metabolismo , Hipotálamo/metabolismo , Orexinas/metabolismo , ARN Nucleolar Pequeño/genética , Sueño/fisiología , Animales , Modelos Animales de Enfermedad , Conducta Alimentaria , Área Hipotalámica Lateral/fisiopatología , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Ratones , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatología
5.
Brain Behav ; 8(12): e01102, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30353709

RESUMEN

INTRODUCTION: In Prader-Willi syndrome (PWS), nonprotein coding small nucleolar (sno) RNAs are involved in the paternally deleted region of chromosome 15q11.2-q13, which is believed to cause the hyperphagic phenotype of PWS. Central to this is SnoRNA116. The supplement Caralluma fimbriata extract (CFE) has been shown to decrease appetite behavior in some individuals with PWS. We therefore investigated the mechanism underpinning the effect of CFE on food intake in the Snord116del mouse. Experiments utilized appetite stimulants which included a 5-hydroxytryptamine (5-HT) 2c receptor antagonist (SB242084), as the 5-HT2cR is implicated in central signaling of satiety. METHODS: After 9-week chronic CFE treatment (33 mg or 100 mg kg-1  day-1 ) or placebo, the 14-week-old Snord116del (SNO) and wild-type mice (n = 72) were rotated through intraperitoneal injections of (a) isotonic saline; (b) 400 mg/kg of 2-deoxyglucose (2DG) (glucose deprivation); (c) 100 mglkg beta-mercaptoacetate (MA), fatty acid signaling; and (d) SB242084 (a selective 5HT2cR antagonist), with 5 days between reagents. Assessments of food intake were from baseline to 4 hr, followed by immunohistochemistry of neural activity utilizing c-Fos, neuropeptide Y, and alpha-melanocyte-stimulating hormone within hypothalamic appetite pathways. RESULTS: Caralluma fimbriata extract administration decreased food intake more strongly in the SNO100CFE group with significantly stimulated food intake demonstrated during coadministration with SB242084. Though stimulatory deprivation was expected to stimulate food intake, 2DG and MA resulted in lower intake in the snord116del mice compared to the WT animals (p = <0.001). Immunohistochemical mapping of hypothalamic neural activity was consistent with the behavioral studies. CONCLUSIONS: This study identifies a role for the 5-HT2cR in CFE-induced appetite suppression and significant stimulatory feeding disruptions in the snord116del mouse model.


Asunto(s)
Apocynaceae , Extractos Vegetales/farmacología , Síndrome de Prader-Willi/tratamiento farmacológico , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Aminopiridinas/farmacología , Animales , Depresores del Apetito/farmacología , Deleción Cromosómica , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Femenino , Eliminación de Gen , Humanos , Hipotálamo/metabolismo , Indoles/farmacología , Masculino , Ratones Endogámicos C57BL , Fenotipo , Fitoterapia , ARN Nucleolar Pequeño/genética , Distribución Aleatoria , Antagonistas del Receptor de Serotonina 5-HT2/farmacología
6.
Nat Commun ; 9(1): 1616, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29691382

RESUMEN

Rhythmic oscillations of physiological processes depend on integrating the circadian clock and diurnal environment. DNA methylation is epigenetically responsive to daily rhythms, as a subset of CpG dinucleotides in brain exhibit diurnal rhythmic methylation. Here, we show a major genetic effect on rhythmic methylation in a mouse Snord116 deletion model of the imprinted disorder Prader-Willi syndrome (PWS). More than 23,000 diurnally rhythmic CpGs are identified in wild-type cortex, with nearly all lost or phase-shifted in PWS. Circadian dysregulation of a second imprinted Snord cluster at the Temple/Kagami-Ogata syndrome locus is observed at the level of methylation, transcription, and chromatin, providing mechanistic evidence of cross-talk. Genes identified by diurnal epigenetic changes in PWS mice overlapped rhythmic and PWS-specific genes in human brain and are enriched for PWS-relevant phenotypes and pathways. These results support the proposed evolutionary relationship between imprinting and sleep, and suggest possible chronotherapy in the treatment of PWS and related disorders.


Asunto(s)
Encéfalo/fisiología , Corteza Cerebral/metabolismo , Ritmo Circadiano , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Síndrome de Prader-Willi/metabolismo
7.
J Clin Invest ; 128(3): 960-969, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29376887

RESUMEN

Profound hyperphagia is a major disabling feature of Prader-Willi syndrome (PWS). Characterization of the mechanisms that underlie PWS-associated hyperphagia has been slowed by the paucity of animal models with increased food intake or obesity. Mice with a microdeletion encompassing the Snord116 cluster of noncoding RNAs encoded within the Prader-Willi minimal deletion critical region have previously been reported to show growth retardation and hyperphagia. Here, consistent with previous reports, we observed growth retardation in Snord116+/-P mice with a congenital paternal Snord116 deletion. However, these mice neither displayed increased food intake nor had reduced hypothalamic expression of the proprotein convertase 1 gene PCSK1 or its upstream regulator NHLH2, which have recently been suggested to be key mediators of PWS pathogenesis. Specifically, we disrupted Snord116 expression in the mediobasal hypothalamus in Snord116fl mice via bilateral stereotaxic injections of a Cre-expressing adeno-associated virus (AAV). While the Cre-injected mice had no change in measured energy expenditure, they became hyperphagic between 9 and 10 weeks after injection, with a subset of animals developing marked obesity. In conclusion, we show that selective disruption of Snord116 expression in the mediobasal hypothalamus models the hyperphagia of PWS.


Asunto(s)
Hiperfagia/metabolismo , Hipotálamo/metabolismo , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño/genética , Animales , Composición Corporal , Dependovirus , Modelos Animales de Enfermedad , Eliminación de Gen , Genotipo , Hiperfagia/genética , Masculino , Ratones , Ratones Transgénicos , Obesidad/metabolismo , Síndrome de Prader-Willi/metabolismo
8.
J Clin Invest ; 128(3): 900-902, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29376891

RESUMEN

Hyperphagia and obesity are the best-known manifestations of Prader-Willi syndrome (PWS) and are responsible for most of the overall morbidity and mortality associated with the disease. Yet these PWS symptoms remain poorly understood and without effective pharmacologic therapies. Mouse models attempting to recapitulate both the genetic alterations and marked hyperphagia plus obesity of PWS have been enigmatic, leading to skepticism about the use of mouse models to investigate PWS. In this issue of the JCI, Polex-Wolf and colleagues challenge the skeptics by successfully inducing hyperphagia following bilateral mediobasal hypothalamic deletion of the Snord116 gene from adult mice. Obesity also resulted, although only in a subset of mice. While this approach represents an exciting advance, highlighting a pathologic effect of loss of mediobasal hypothalamic Snord116 expression on the development of PWS's hallmark symptoms, the variability in the body-weight and body composition responses to this site-selective gene deletion raises several questions.


Asunto(s)
Síndrome de Prader-Willi , Animales , Hiperfagia , Hipotálamo , Ratones , Obesidad , ARN Nucleolar Pequeño
9.
J Neuroendocrinol ; 29(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28094877

RESUMEN

The Snord116 gene cluster has been recognised as a critical contributor to the Prader-Willi syndrome (PWS), with mice lacking Snord116 displaying many classical PWS phenotypes, including low postnatal body weight, reduced bone mass and increased food intake. However, these mice do not develop obesity as a result of increased energy expenditure. To understand the physiological function of SNORD116 better and potentially rescue the altered metabolism of Snord116-/- mice, we used an adeno-associated viral (AAV) approach to reintroduce the product of the Snord116 gene into the hypothalamus in Snord116-/- mice at different ages. The results obtained show that mid-hypothalamic re-introduction of SNORD116 in 6-week-old Snord116-/- mice leads to significantly reduced body weight and weight gain, which is associated with elevated energy expenditure. Importantly, when the intervention targets other areas such as the anterior region of the hypothalamus or the reintroduction occurs in older mice, the positive effects on energy expenditure are diminished. These data indicate that the metabolic symptoms of PWS develop gradually and the Snord116 gene plays a critical role during this process. Furthermore, when we investigated the consequences of SNORD116 re-introduction under conditions of thermoneutrality where the mild cold stress influences are avoided, we also observed a significant increase in energy expenditure. In conclusion, the rescue of mid-hypothalamic Snord116 deficiency in young Snord116 germline deletion mice increases energy expenditure, providing fundamental information contributing to potential virus-mediated genetic therapy in PWS.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Síndrome de Prader-Willi/metabolismo , ARN Nucleolar Pequeño/metabolismo , Animales , Peso Corporal , Masculino , Ratones , Ratones Noqueados , ARN Nucleolar Pequeño/genética
10.
Neuropeptides ; 61: 87-93, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27823858

RESUMEN

Germline deletion of the Prader-Willi syndrome (PWS) candidate gene Snord116 in mice leads to some classical symptoms of human PWS, notably reductions in body weight, linear growth and bone mass. However, Snord116 deficient mice (Snord116-/-) do not develop an obese phenotype despite their increased food intake and the underlying mechanism for that is unknown. We tested the phenotypes of germline Snord116-/- as well as neuropeptide Y (NPY) neuron specific Snord116lox/lox/NPYcre/+ mice at 30°C, the thermoneutral temperature of mice, and compared these to previous reports studies conducted at normal room temperature. Snord116-/- mice at 30°C still weighed less than wild type but had increased body weight gain. Importantly, food intake and energy expenditure were no longer different at 30°C, and the reduced bone mass and nasal-anal length observed in Snord116-/- mice at room temperature were also normalized. Mechanistically, the thermoneutral condition led to the correction of the mRNA expression of NPY and pro-opiomelanocortin (POMC), which were both previously observed to be significantly up-regulated at room temperature. Importantly, almost identical phenotypes and NPY/POMC mRNA expression alterations were also observed in Snord116lox/lox/NPYcre/+ mice, which lack the Snord116 gene only in NPY neurons. These data illustrate that mild cold stress is a critical factor preventing the development of obesity in Snord116-/- mice via the NPY system. Our study highlights that the function of Snord116 in the hypothalamus may be to enhance energy expenditure, likely via the NPY system, and also indicates that Snord116 function in mice is strongly dependent on environmental conditions such as cold exposure.


Asunto(s)
Metabolismo Energético/genética , Homeostasis/genética , Neuronas/metabolismo , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño/genética , Animales , Peso Corporal/genética , Ingestión de Alimentos/genética , Hipotálamo/metabolismo , Ratones , Ratones Noqueados , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Síndrome de Prader-Willi/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , ARN Nucleolar Pequeño/metabolismo , Temperatura
11.
Thromb Haemost ; 115(6): 1147-56, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26940969

RESUMEN

Multiple studies have shown the effects of long-term exposure to high-fat or western diets on the vascular system. There is limited knowledge on the acute effects of high circulating fat levels, specifically on platelets, which have a role in many processes, including thrombosis and inflammation. This study investigated the effects of acute, high-fat exposure on platelet function and transcript profile. Twenty healthy participants were given an intravenous infusion of 20% Intralipid emulsion and heparin over 6 hours. Blood samples were taken prior to and the day after infusion to measure platelet function and transcript expression levels. Platelet aggregation was not significantly affected by Intralipid infusion, but, when mitochondria function was inhibited by carbonyl cyanide 3-chlorophenylhydrazone (CCCP) or oligomycin, platelet aggregation was higher in the post-infusion state compared to baseline. Through RNA sequencing, and verified by RT-qPCR, 902 miRNAs and 617 mRNAs were affected by Intralipid infusion. MicroRNAs increased include miR-4259 and miR-346, while miR-517b and miR-517c are both decreased. Pathway analysis identified two clusters significantly enriched, including cell motility. In conclusion, acute exposure to high fat affects mitochondrial-dependent platelet function, as well as the transcript profile.


Asunto(s)
Plaquetas/metabolismo , Fosfolípidos/administración & dosificación , Aceite de Soja/administración & dosificación , Adulto , Anciano , Dieta Alta en Grasa/efectos adversos , Emulsiones/administración & dosificación , Femenino , Voluntarios Sanos , Humanos , Infusiones Intravenosas , Masculino , MicroARNs/sangre , MicroARNs/genética , Persona de Mediana Edad , Agregación Plaquetaria , ARN Mensajero/sangre , ARN Mensajero/genética , ARN Nucleolar Pequeño/sangre , ARN Nucleolar Pequeño/genética , Transcripción Genética , Adulto Joven
12.
Sci Rep ; 6: 18614, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26726071

RESUMEN

Prader-Willi syndrome (PWS) is the predominant genetic cause of obesity in humans. Recent clinical reports have suggested that micro-deletion of the Snord116 gene cluster can lead to PWS, however, the extent of the contributions of the encoded snoRNAs is unknown. Here we show that mice lacking Snord116 globally have low birth weight, increased body weight gain, energy expenditure and hyperphagia. Consistent with this, microarray analysis of hypothalamic gene expression revealed a significant alteration in feeding related pathways that was also confirmed by in situ hybridisation. Importantly, selective deletion of Snord116 only from NPY expressing neurons mimics almost exactly the global deletion phenotype including the persistent low birth weight, increased body weight gain in early adulthood, increased energy expenditure and hyperphagia. Mechanistically, the lack of Snord116 in NPY neurons leads to the upregulation of NPY mRNA consistent with the hyperphagic phenotype and suggests a critical role of Snord116 in the control of NPY neuronal functions that might be dysregulated in PWS.


Asunto(s)
Regulación del Apetito , ARN Nucleolar Pequeño/fisiología , Animales , Composición Corporal , Peso Corporal , Metabolismo de los Hidratos de Carbono , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos , Metabolismo Energético , Femenino , Expresión Génica , Hipotálamo/metabolismo , Masculino , Ratones Noqueados , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Obesidad/etiología , Obesidad/genética
13.
Gene ; 572(2): 266-73, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26220404

RESUMEN

The loss of two gene clusters encoding small nucleolar RNAs, SNORD115 and SNORD116 contribute to Prader-Willi syndrome (PWS), the most common syndromic form of obesity in humans. SNORD115 and SNORD116 are considered to be orphan C/D box snoRNAs (SNORDs) as they do not target rRNAs or snRNAs. SNORD115 exhibits sequence complementarity towards the serotonin receptor 2C, but SNORD116 shows no extended complementarities to known RNAs. To identify molecular targets, we performed genome-wide array analysis after overexpressing SNORD115 and SNORD116 in HEK 293T cells, either alone or together. We found that SNORD116 changes the expression of over 200 genes. SNORD116 mainly changed mRNA expression levels. Surprisingly, we found that SNORD115 changes SNORD116's influence on gene expression. In similar experiments, we compared gene expression in post-mortem hypothalamus between individuals with PWS and aged-matched controls. The synopsis of these experiments resulted in 23 genes whose expression levels were influenced by SNORD116. Together our results indicate that SNORD115 and SNORD116 influence expression levels of multiple genes and modify each other activity.


Asunto(s)
Regulación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Células HEK293 , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Familia de Multigenes , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/patología , ARN Mensajero/genética
14.
Int J Dev Neurosci ; 30(6): 479-85, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22664655

RESUMEN

The hypothalamus is integral to the regulation of body homeostasis, including food intake, energy balance, and blood pressure. Dysfunction of the hypothalamus has been associated with a broad range of disorders; many of which are sex-dependent in prevalence. Small nucleolar (sno) RNAs are a group of small RNAs located in nucleoli that modulate chemical modifications and maturation of ribosomal or other RNAs. Recent data suggest that snoRNA Snord116 is important for the pathogenesis of Prader-Willi syndrome (PWS) characterized by hyperphagia and obesity. The current study was conducted to assess a potential cellular link between Snord116 and phenotypes of PWS. Data from mice revealed Snord116 expression in the medial hypothalamus, particularly within nuclei that are part of feeding circuitry. High expression of Snord116 was evident in the paraventricular (PVN) and ventromedial (VMH) nuclei, but particularly prevalent in the arcuate nucleus (ARC) according to in situ hybridization. Snord116 expression level in ventral hypothalamic dissections including ARC was significantly greater (by 2-fold) than that in cortex; and its expression level in dorsal hypothalamic dissections including PVN was double that in cortex. The enhanced expression pattern of Snord116 in hypothalamic nuclei was observed at weaning and young adult stages, but less obvious neonatally when expression was significantly more widespread. Therefore the expression of Snord116 likely is regulated developmentally. These results provide a new lead for understanding the mechanism(s) related to hyperphagia and obesity symptoms in PWS patients.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Hipotálamo/anatomía & histología , Hipotálamo/metabolismo , ARN Nucleolar Pequeño/metabolismo , Animales , Animales Recién Nacidos , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Embrión de Mamíferos , Femenino , Hipotálamo/embriología , Hipotálamo/crecimiento & desarrollo , Masculino , Ratones , ARN Nucleolar Pequeño/genética
15.
PLoS One ; 5(7): e11779, 2010 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-20668672

RESUMEN

Non-coding RNAs (ncRNAs) are an essential class of molecular species that have been difficult to monitor on high throughput platforms due to frequent lack of polyadenylation. Using a polyadenylation-neutral amplification protocol and next-generation sequencing, we explore ncRNA expression in eleven human tissues. ncRNAs 7SL, U2, 7SK, and HBII-52 are expressed at levels far exceeding mRNAs. C/D and H/ACA box snoRNAs are associated with rRNA methylation and pseudouridylation, respectively: spleen expresses both, hypothalamus expresses mainly C/D box snoRNAs, and testes show enriched expression of both H/ACA box snoRNAs and RNA telomerase TERC. Within the snoRNA 14q cluster, 14q(I-6) is expressed at much higher levels than other cluster members. More reads align to mitochondrial than nuclear tRNAs. Many lincRNAs are actively transcribed, particularly those overlapping known ncRNAs. Within the Prader-Willi syndrome loci, the snoRNA HBII-85 (group I) cluster is highly expressed in hypothalamus, greater than in other tissues and greater than group II or III. Additionally, within the disease locus we find novel transcription across a 400,000 nt span in ovaries. This genome-wide polyA-neutral expression compendium demonstrates the richness of ncRNA expression, their high expression patterns, their function-specific expression patterns, and is publicly available.


Asunto(s)
Genoma Humano/genética , ARN Nucleolar Pequeño/genética , ARN no Traducido/genética , Perfilación de la Expresión Génica , Humanos , Hipotálamo/metabolismo , Reacción en Cadena de la Polimerasa , Bazo/metabolismo
16.
PLoS One ; 5(2): e9402, 2010 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-20195375

RESUMEN

Neonatal feeding problems are observed in several genetic diseases including Prader-Willi syndrome (PWS). Later in life, individuals with PWS develop hyperphagia and obesity due to lack of appetite control. We hypothesized that failure to thrive in infancy and later-onset hyperphagia are related and could be due to a defect in the hypothalamus. In this study, we performed gene expression microarray analysis of the hypothalamic response to maternal deprivation in neonatal wild-type and Snord116del mice, a mouse model for PWS in which a cluster of imprinted C/D box snoRNAs is deleted. The neonatal starvation response in both strains was dramatically different from that reported in adult rodents. Genes that are affected by adult starvation showed no expression change in the hypothalamus of 5 day-old pups after 6 hours of maternal deprivation. Unlike in adult rodents, expression levels of Nanos2 and Pdk4 were increased, and those of Pgpep1, Ndp, Brms1l, Mett10d, and Snx1 were decreased after neonatal deprivation. In addition, we compared hypothalamic gene expression profiles at postnatal days 5 and 13 and observed significant developmental changes. Notably, the gene expression profiles of Snord116del deletion mice and wild-type littermates were very similar at all time points and conditions, arguing against a role of Snord116 in feeding regulation in the neonatal period.


Asunto(s)
Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hipotálamo/metabolismo , Privación Materna , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Privación de Alimentos , Genotipo , Humanos , Hipotálamo/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
BMC Evol Biol ; 7: 34, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17343755

RESUMEN

BACKGROUND: Tnt1 was the first active plant retrotransposon identified in tobacco after nitrate reductase gene disruption. The Tnt1 superfamily comprises elements from Nicotiana (Tnt1 and Tto1) and Lycopersicon (Retrolyc1 and Tlc1) species. The study presented here was conducted to characterise Tnt1-related sequences in 20 wild species of Solanum and five cultivars of Solanum tuberosum. RESULTS: Tnt1-related sequences were amplified from total genomic DNA using a PCR-based approach. Purified fragments were cloned and sequenced, and clustering analysis revealed three groups that differ in their U3 region. Using a network approach with a total of 453 non-redundant sequences isolated from Solanum (197), Nicotiana (140) and Lycopersicon (116) species, it is demonstrated that the Tnt1 superfamily can be treated as a population to resolve previous phylogenetic multifurcations. The resulting RNAseH network revealed that sequences group according to the Solanaceae genus, supporting a strong association with the host genome, whereas tracing the U3 region sequence association characterises the modular evolutionary pattern within the Tnt1 superfamily. Within each genus, and irrespective of species, nearly 20% of Tnt1 sequences analysed are identical, indicative of being part of an active copy. The network approach enabled the identification of putative "master" sequences and provided evidence that within a genus these master sequences are associated with distinct U3 regions. CONCLUSION: The results presented here support the hypothesis that the Tnt1 superfamily was present early in the evolution of Solanaceae. The evidence also suggests that the RNAseH region of Tnt1 became fixed at the host genus level whereas, within each genus, propagation was ensured by the diversification of the U3 region. Different selection pressures seemed to have acted on the U3 and RNAseH modules of ancestral Tnt1 elements, probably due to the distinct functions of these regions in the retrotransposon life cycle, resulting in both co evolution and adaptation of the element population with its host.


Asunto(s)
Genes de Plantas , Filogenia , Retroelementos , Solanum/genética , Secuencia de Bases , Evolución Molecular , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Nucleolar Pequeño/genética , Ribonucleasa H/genética , Alineación de Secuencia , Especificidad de la Especie , Secuencias Repetidas Terminales/genética
18.
J Biol Chem ; 280(6): 4021-4, 2005 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-15590684

RESUMEN

The 2,2,7-trimethylguanosine (TMG) cap structure is characteristic of certain eukaryotic small nuclear and small nucleolar RNAs. Prior studies have suggested that cap trimethylation might be contingent on cis-acting elements in the RNA substrate, protein components of a ribonucleoprotein complex, or intracellular localization of the RNA substrate. However, the enzymatic requirements for TMG cap formation remain obscure because TMG synthesis has not been reconstituted in vitro from defined components. Tgs1 is a conserved eukaryal protein that was initially identified as being required for RNA cap trimethylation in vivo in budding yeast. Here we show that purified recombinant fission yeast Tgs1 catalyzes methyl transfer from S-adenosylmethionine (AdoMet) to m7GTP and m7GDP. Tgs1 also methylates the cap analog m(7)GpppA but is unreactive with GTP, GDP, GpppA, m2,2,7GTP, m2,2,7GDP, ATP, CTP, UTP, and ITP. The products of methyl transfer to m7GTP and m7GDP formed under conditions of excess methyl acceptor are 2,7-dimethyl GTP and 2,7-dimethyl GDP, respectively. Under conditions of limiting methyl acceptor, the initial m2,7GDP product is converted to m2,2,7GDP in the presence of excess AdoMet. We conclude that Tgs1 is guanine-specific, that N7 methylation must precede N2 methylation, that Tgs1 acts via a distributive mechanism, and that the chemical steps of TMG synthesis do not require input from RNA or protein cofactors.


Asunto(s)
Guanosina/análogos & derivados , Metiltransferasas/química , Metiltransferasas/fisiología , Cromatografía en Capa Delgada , Metilación de ADN , ADN Complementario/metabolismo , Ditiotreitol/farmacología , Relación Dosis-Respuesta a Droga , Guanina/química , Guanosina/química , Guanosina Difosfato/química , Metilación , Unión Proteica , ARN/química , ARN Nucleolar Pequeño/química , Proteínas Recombinantes/química , S-Adenosilmetionina/química , Schizosaccharomyces/metabolismo , Factores de Tiempo
19.
Trends Cell Biol ; 12(2): 90-6, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11849973

RESUMEN

Recent studies of the eukaryotic ribosomal RNA processing pathway have identified a complex of ten riboexonucleases called the exosome that plays a central role in the precise formation of the 3' ends of several types of RNAs. The exosome also destroys excess ribosomal RNA precursors and unused intermediates and degrades poly(A)-mRNAs in the cytoplasm. In the nucleus, the complex appears to function in a regulated mRNA surveillance system that degrades transcripts in response to defects in the mRNA processing and export pathways. How the cell regulates the nucleolytic prowess of the exosome to ensure correct and timely synthesis and destruction of RNAs is a central focus of current research.


Asunto(s)
Exorribonucleasas/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN Ribosómico/metabolismo , Animales , Humanos , ARN Nuclear Pequeño/metabolismo , ARN Nucleolar Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA