Добірка наукової літератури з теми "SNORD115"

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся зі списками актуальних статей, книг, дисертацій, тез та інших наукових джерел на тему "SNORD115".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Статті в журналах з теми "SNORD115":

1

Falaleeva, Marina, Carlos R. Sulsona, Horst R. Zielke, Kathleen M. Currey, Pierre de la Grange, Vahid Aslanzadeh, Daniel J. Driscoll, and Stefan Stamm. "Molecular Characterization of a Patient Presumed to Have Prader-Willi Syndrome." Clinical Medicine Insights: Case Reports 6 (January 2013): CCRep.S11510. http://dx.doi.org/10.4137/ccrep.s11510.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Prader-Willi syndrome (PWS) is caused by the loss of RNA expression from an imprinted region on chromosome 15 that includes SNRPN, SNORD115, and SNORD116. Currently, there are no mouse models that faithfully reflect the human phenotype and investigations rely on human post-mortem material. During molecular characterization of tissue deposited in a public brain bank from a patient diagnosed with Prader-Willi syndrome, we found RNA expression from SNRPN, SNORD115, and SNORD116 which does not support a genetic diagnosis of Prader-Willi syndrome. The patient was a female, Caucasian nursing home resident with history of morbid obesity (BMI 56.3) and mental retardation. She died at age of 56 from pulmonary embolism. SNORD115 and SNORD116 are unexpectedly stable in post mortem tissue and can be used for post-mortem diagnosis. Molecular characterization of PWS tissue donors can confirm the diagnosis and identify those patients that have been misdiagnosed.
2

Falaleeva, Marina, Justin Surface, Manli Shen, Pierre de la Grange, and Stefan Stamm. "SNORD116 and SNORD115 change expression of multiple genes and modify each other's activity." Gene 572, no. 2 (November 2015): 266–73. http://dx.doi.org/10.1016/j.gene.2015.07.023.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

DE ROO, A., B. FOETS, and J. VAN DEN OORD. "FRO 2014: The pathophysiologic role of SNORD115 and SNORD116 in late-onset Fuchs' endothelial corneal dystrophy." Acta Ophthalmologica 92 (August 20, 2014): 0. http://dx.doi.org/10.1111/j.1755-3768.2014.4663.x.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Ronchetti, Domenica, Katia Todoerti, Giacomo Tuana, Luca Agnelli, Laura Mosca, Marta Lionetti, Sonia Fabris, et al. "The Expression Pattern of Small Nucleolar and Small Cajal Body-Specific RNAs Characterizes Distinct Molecular Subtypes of Multiple Myeloma." Blood 120, no. 21 (November 16, 2012): 3955. http://dx.doi.org/10.1182/blood.v120.21.3955.3955.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Abstract 3955 Small nucleolar RNAs (snoRNAs) and small Cajal body-specific RNAs (scaRNAs) are non-coding RNAs involved in the maturation of other RNA molecules and generally located in the introns of host genes. It is an emerging evidence that altered sno/scaRNAs expression may play a pathological role in cancer. Impaired sno/scaRNAs expression has recently been reported both in acute leukemia and smoldering myeloma that rapidly progressed to symptomatic disease. In addition, as regards multiple myeloma (MM), very recent data suggested an oncogenic role for SCARNA22 in those MM patients over-expressing SCARNA22/MMSET as a result of t(4;14) translocation. However, comprehensive information concerning the expression behavior of sno/scaRNAs in MM is still lacking. This study elucidates the patterns of sno/scaRNAs expression in MM by profiling purified malignant plasma cells from 55 MMs, 8 secondary plasma cell leukemias (sPCL) and 4 normal controls using Human Gene 1.0 ST arrays. Overall, a global sno/scaRNAs down-regulation was found in MMs and at more extent in sPCLs compared to normal plasma cells. Whereas SCARNA22 resulted the only sno/scaRNA characterizing the TC4 MM, TC2 group displayed a distinct sno/scaRNA signature overexpressing members of SNORD115 and SNORD116 families located in a region finely regulated by an imprinting center at 15q11 which, however, resulted overall hypomethylated in MMs independently of the SNORD115 and SNORD116 expression levels. In addition, impaired expression of sno/scaRNAs raised from the comparison between MM and sPCL, suggested a role in tumor progression. Furthermore, to uncover possible mechanisms at the basis of sno/scaRNAs deregulation, we investigated the correlation between sno/scaRNAs and the corresponding host-genes expression levels, outlining the coordinated expression of up to 50% of sno/scaRNAs/host-genes pairs. Finally, we investigated whether the sno/scaRNAs transcriptional pattern may be influenced by allelic imbalances involving their genomic location, as already demonstrated concerning mRNA expression, and revealed a dosage effect involving several chromosomal regions. Our data extend the current view of sno/scaRNAs deregulation in cancer and add novel information into the bio-molecular complexity of plasma cell dyscrasias. Furthermore, our findings may contribute to develop functional approaches to examine the activity of deregulated sno/scaRNAs in MM, as well as to further enlighten their possible role as targets of novel therapeutic agents. Disclosures: No relevant conflicts of interest to declare.
5

Glatt-Deeley, Heather, Daria L. Bancescu, and Marc Lalande. "Prader–Willi syndrome, Snord115, and Htr2c editing." neurogenetics 11, no. 1 (August 4, 2009): 143–44. http://dx.doi.org/10.1007/s10048-009-0209-x.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Bortolin-Cavaille, M. L., and J. Cavaille. "The SNORD115 (H/MBII-52) and SNORD116 (H/MBII-85) gene clusters at the imprinted Prader-Willi locus generate canonical box C/D snoRNAs." Nucleic Acids Research 40, no. 14 (April 11, 2012): 6800–6807. http://dx.doi.org/10.1093/nar/gks321.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Davies, Jennifer R., Lawrence S. Wilkinson, Anthony R. Isles, and Trevor Humby. "Prader–Willi syndrome imprinting centre deletion mice have impaired baseline and 5-HT2CR-mediated response inhibition." Human Molecular Genetics 28, no. 18 (May 14, 2019): 3013–23. http://dx.doi.org/10.1093/hmg/ddz100.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Prader–Willi syndrome (PWS) is a neurodevelopmental disorder caused by deletion or inactivation of paternally expressed imprinted genes on human chromosome 15q11–q13. In addition to endocrine and developmental issues, PWS presents with behavioural problems including stereotyped behaviour, impulsiveness and cognitive deficits. The PWS genetic interval contains several brain-expressed small nucleolar (sno) RNA species that are subject to genomic imprinting, including snord115 that negatively regulates post-transcriptional modification of the serotonin 2C receptor (5-HT2CR) pre-mRNA potentially leading to a reduction in 5-HT2CR function. Using the imprinting centre deletion mouse model for PWS (PWSICdel) we have previously shown impairments in a number of behaviours, some of which are abnormally sensitive to 5-HT2CR-selective drugs. In the stop-signal reaction time task test of impulsivity, PWSICdel mice showed increased impulsivity relative to wild-type (WT) littermates. Challenge with the selective 5-HT2CR agonist WAY163909 reduced impulsivity in PWSICdel mice but had no effect on WT behaviour. This behavioural dissociation in was also reflected in differential patterns of immunoreactivity of the immediate early gene c-Fos, with a blunted response to the drug in the orbitofrontal cortex of PWSICdel mice, but no difference in c-Fos activation in the nucleus accumbens. These findings suggest specific facets of response inhibition are impaired in PWSICdel mice and that abnormal 5-HT2CR function may mediate this dissociation. These data have implications for our understanding of the aetiology of PWS-related behavioural traits and translational relevance for individuals with PWS who may seek to control appetite with the new obesity treatment 5-HT2CR agonist lorcaserin.
8

Kocher, Matthew A., Fenix W. Huang, Erin Le, and Deborah J. Good. "Snord116 Post-transcriptionally Increases Nhlh2 mRNA Stability: Implications for Human Prader-Willi Syndrome." Human Molecular Genetics 30, no. 12 (April 15, 2021): 1101–10. http://dx.doi.org/10.1093/hmg/ddab103.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract 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.
9

Langouët, Maéva, Dea Gorka, Clarisse Orniacki, Clémence M. Dupont-Thibert, Michael S. Chung, Heather R. Glatt-Deeley, Noelle Germain, et al. "Specific ZNF274 binding interference at SNORD116 activates the maternal transcripts in Prader-Willi syndrome neurons." Human Molecular Genetics 29, no. 19 (September 25, 2020): 3285–95. http://dx.doi.org/10.1093/hmg/ddaa210.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Prader-Willi syndrome (PWS) is characterized by neonatal hypotonia, developmental delay and hyperphagia/obesity. This disorder is caused by the absence of paternally expressed gene products from chromosome 15q11–q13. We previously demonstrated that knocking out ZNF274, a Kruppel-associated box-A-domain zinc finger protein capable of recruiting epigenetic machinery to deposit the H3K9me3 repressive histone modification, can activate expression from the normally silent maternal allele of SNORD116 in neurons derived from PWS induced pluripotent stem cells (iPSCs). However, ZNF274 has many other targets in the genome in addition to SNORD116. Depleting ZNF274 will surely affect the expression of other important genes and disrupt other pathways. Here, we used CRISPR/Cas9 to delete ZNF274 binding sites at the SNORD116 locus to determine whether activation of the maternal copy of SNORD116 could be achieved without altering ZNF274 protein levels. We obtained similar activation of gene expression from the normally silenced maternal allele in neurons derived from PWS iPSCs, compared with ZNF274 knockout, demonstrating that ZNF274 is directly involved in the repression of SNORD116. These results suggest that interfering with ZNF274 binding at the maternal SNORD116 locus is a potential therapeutic strategy for PWS.
10

Zhang, Cong, Lian-mei Zhao, Hao Wu, Guo Tian, Su-li Dai, Ri-yang Zhao, and Bao-en Shan. "C/D-Box Snord105b Promotes Tumorigenesis in Gastric Cancer via ALDOA/C-Myc Pathway." Cellular Physiology and Biochemistry 45, no. 6 (2018): 2471–82. http://dx.doi.org/10.1159/000488265.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Background/Aims: Small nucleolar RNAs (snoRNAs) play an important role in carcinogenesis. In this study, we identified a C/D box snoRNA, snord105b, and further investigated the function and mechanism of the snord105b in gastric cancer (GC). Methods: The expression level of snord105b in GC tissures, sera and cell lines were detected by qRT-PCR. Cell viability was assessed using MTS assay. Transwell and wound healing assay were performed to evaluate migration and invasion, and protein expression was examined by western blotting. ChIRP and MS analysis was used to seek for the special binding protein of snord105b. Results: The snord105b was upregulated and associated with tumor size, differentiation, and pathological stage in GC. Snord105b affected proliferation, migration and invasion in multiple GC cell lines. The oncoqenic activity of snord105b was also confirmed with in vivo data. Mechanistically, snord105b specifically bound to ALDOA and affected C-myc, which plays a key role in carcinogenesis and tumor development. Conclusion: Snord105b appears to be a novel oncogene and is clinically and functionally involved in the development of GC. Targeting snord105b and its pathway may provide new biomarkers or potential treatments for patients with GC.

Дисертації з теми "SNORD115":

1

Hebras, Jade. "Caractérisation moléculaire du petit ARN nucléolaire SNORD115 : un rôle dans la régulation de l'expression et de la fonction du récepteur à la sérotonine 5-HT2C ?" Thesis, Toulouse 3, 2020. http://www.theses.fr/2020TOU30209.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Le nucléole des mammifères contient des centaines de petits ARN nucléolaires à boîte C/D (SNORD) dont la grande majorité guide une 2'-O-ribose méthylation sur les précurseurs des ARN ribosomiques (pré-ARNr). Certains SNORD facilitent aussi les clivages que subissent le pré-ARNr ou modifient le petit ARN nucléaire U6. Des travaux récents laissent également entrevoir que certains SNORD interagissent avec des ARNm. C'est le cas par exemple pour SNORD115 qui est au cœur de mon travail de thèse. SNORD115 est exprimé uniquement dans le cerveau à partir de nombreux gènes répétés en tandem situés au locus SNURF-SNRPN dont l'expression est contrôlée par l'empreinte génomique parentale. Des défauts génétiques associés à ce locus chromosomique sont associés à une maladie rare: le syndrome de Prader-Willi (SPW). SNORD115 est remarquable car il possède une longue complémentarité conservée avec l'ARNm codant un récepteur à la sérotonine, le variant 5-HT2C. Certains travaux proposent que SNORD115 régule la voie 5-HT2C en modulant l'épissage alternatif ou l'édition A vers I du pré-ARNm 5-HT2C. Un défaut dans l'activité du 5-HT2C pourrait être à l'origine de l'hyperphagie et/ou des anomalies comportementales qui caractérisent le SPW. Mon projet de thèse principal consistait à éprouver cette hypothèse grâce à un nouveau modèle murin CRISPR/Cas9 invalidé pour SNORD115. Mes résultats montrent que la perte d'expression de SNORD115 ne perturbe pas la régulation post-transcriptionnelle du pré-ARNm 5-HT2C in vivo. D'autre part, des études réalisées dans l'équipe n'ont pas permis de révéler des anomalies marquées dans les phénotypes anxio-dépressifs, ni dans le comportement alimentaire. Ma thèse soulève donc des questions importantes quant au rôle régulateur de SNORD115 dans le cerveau et de sa contribution potentielle dans l'étiologie du SPW. En parallèle, j'ai aussi abordé le répertoire des 2'-O-méthylations de l'ARNr dans des tissus murins, notamment le cerveau. Ce travail s'inscrivait dans la thématique émergente de la théorie du "ribosome spécialisé" qui propose qu'une hétérogénéité structurale des composants du ribosome puisse se traduire par des changements dans les capacités fonctionnelles du ribosome. Mes résultats montrent des variations dans la méthylation pour un nombre très limité de sites, et ce principalement au cours du développement. Aussi, les ribosomes des tissus développementaux sont globalement moins méthylés que ceux des tissus adultes. Nous avons concentré nos efforts sur LSU-G4593 dont la méthylation guidée par SNORD78 est retrouvée uniquement au cours du développement. Nous proposons que des évènements d'épissage alternatif du gène-hôte de SNORD78 modulent la production de SNORD78, et de fait le niveau de méthylation LSU-Gm4593. Grâce à l'étude d'une lignée cellulaire humaine (HEK293) invalidée pour SNORD78, j'ai recherché les implications fonctionnelles de LSU-Gm4593. A ce jour, mes travaux ne montrent pas un rôle marqué dans la prolifération cellulaire, ni dans la fidélité de la traduction. La fonction précise de LSU-Gm4593 demeure donc incomprise
The nucleolus of mammalian cells contains hundreds of box C/D small nucleolar RNAs (SNORDs). Majority of them, guide sequence-specific 2'-O ribose methylations into ribosomal RNA (rRNA). Some of them facilitate RNA folding and cleavages of ribosomal RNA precursors or guide ribose methylations into spliceosomal small nuclear RNA U6. Recent studies propose that some SNORD could target other transcripts, possibly messenger RNA as suggested by the brain-specific SNORD115. SNORD115 is processed from tandemly repeated genes embedded in the imprinted SNURF-SNRPN domain. Defects in gene expression at this domain are causally linked to rare disease: the Prader-Willi Syndrome (PWS). Excitingly, SNORD115 displays an extensive region of complementary to a brain-specific mRNA encoding the serotonin receptor 5-HT2C. SNORD115 could influence 5-HT2C signaling by fine-tuning alternative splicing or A to I RNA editing of 5-HT2C pre-mRNA. Reduced 5-HT2C receptor activity could contribute to impaired emotional response and/or compulsive overeating that characterized the syndrome. My work was to test this hypothesis using a CRISPR/Cas9-mediated SNORD115 knockout mouse model. My results show that loss of SNORD115 expression, in vivo, does not alter the post-transcriptional regulation of 5-HT2C pre-mRNA processing. Others results from the team do not reveal any defects in anxio-depressive phenotypes and eating behaviour. Our study questions the regulatory roles of SNORD115 in brain functions and behavioural disturbance associated with PWS. On other hand, I have studied ribose methylation sites in rRNA from mouse tissues. This work was included in emerging field of the specialized ribosome hypothesis which suggests heterogeneity in ribosomes may impact activity of ribosomes. Our results show significant changes at few discrete set of sites, especially in rRNA from developing tissues. Also, rRNA from developing tissues is globally less methylated than rRNA from adult tissues. We focus on LSU-Gm4593 site because this position is specifically methylated only during development and hardly ever detected in adult tissues. Methylation at LSU-G4593 is guided by SNORD78. We propose that the expression levels of SNORD78 during development appeared to be regulated by alternative splicing of the host-gene and to correlate with the methylation level of its target site at LSU-G4593. We've used a human cell line (HEK293T) inactivated for the SNORD78 gene in order to understand the functionally role of the corresponding ribose methylation. Our work did not demonstrate any overt cellular phenotypes, even though translation fidelity and the precise function of LSU-Gm4593 remains unknown
2

Purmann, Carolin. "The role of SNORD116 in Prader-Willi syndrome." Thesis, University of Cambridge, 2012. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.610424.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Cruvinel, Estela Mitie. "Estudo da expressão diferencial de genes localizados no segmento cromossômico 15q11-q13 em pacientes com as síndromes de Angelman e Prader-Willi." Universidade de São Paulo, 2015. http://www.teses.usp.br/teses/disponiveis/41/41131/tde-24092015-133351/.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
A síndrome de Prader Willi (PWS) é uma doença de neurodesenvolvimento; a principal hipótese de causa de PWS é a ausência da expressão de SNORD116. O SNORD116 fica na região 15q11-q13 que apresenta vários genes com imprinting genômico e é conhecida por ser controlada pela região de controle de imprinting PWS (PWS-IC) que se localiza sobreposta à região promotora e ao exon 1 do gene SNRPN. Em camundongos, uma proteína zinc finger (Zfp57) foi descrita como importante para o estabelecimento e manutenção do imprinting no Snrpn. Através de análise do ENCODE do Genome Browser, verificamos que outra proteína zinc finger (ZNF274) se liga ao SNORD116. ZNF274 é conhecida por formar um complexo com TRIM28 e SETDB1 que inibe a expressão através da trimetilação da lisina 9 na histona 3 (H3K9me3). No atual estudo mostramos que ZNF274 se liga ao SNORD116 preferencialmente ao alelo materno nas células-tronco pluripotente induzidas (iPSCs). Adicionalmente, as proteínas TRIM28 e SETDB1, que formam um complexo com a ZNF274, estão presentes na região do SNORD116, e a modificação H3K9me3 ocorre preferencialmente no alelo materno nas iPSCs. Na análise funcional, mostramos que o knockdown de SETDB1 isoladamente ou combinado com o knockdown de ZNF274 causa aumento na expressão de SNRPN e SNORD116 nas iPSCs. Além disso, ocorre redução do H3K9me3 e aumento da modificação relacionada à ativação da transcrição, H3K4me2 (dimetilação da lisina 4 na histona 3), na PWS-IC. Os knockdowns também afetam a metilação de DNA, ocasionando o aumento de 5-hidroximetliação de citosinas na PWS-IC. Em outros tipos celulares estudados, neurônios derivados de iPSCs e SHEDs, ZNF274 e a modificação H3K9me3 ocorrem em ambos os alelos dentro do SNORD116. É possível que, nas iPSCs, este complexo proteja a região imprintada da desmetilação do DNA de proteína(s) que atue(m) nessa região somente em células pluripotentes. Nossos achados possibilitam melhor compreensão dos mecanismos envolvidos no imprinting da região 15q11-q13, principalmente do SNORD116, e, consequentemente, disponibiliza novas ferramentas para o desenvolvimento de futuras terapias para PWS.
Prader-Willi syndrome (PWS) is a neurodevelopmental disorder. Loss of paternal copies of the cluster of SNORD116 C/D box snoRNAs and their host transcript, 116HG, on human chromosome 15q11-q13 imprinted region is considered to be the major responsible for PWS. PWS-imprinting center (PWS-IC) regulates 15q11-q13 imprinting. PWS-IC is located upstream and in the exon 1 of SNURF-SNRPN gene. In mice, Zfp57 plays an important role in establishment and maintenance of Snrpn imprinting. In human, ENCODE database indicates that ZNF274 binds to SNORD116. Moreover, ZNF274 are C2H2/KRAB zinc finger proteins as Zfp57. We have investigated the mechanism of repression of the maternal SNORD116. Here, we report that the ZNF274, in association with the histone H3 lysine 9 (H3K9) methyltransferase SETDB1, is part of a complex that binds to the silent maternal but not to the active paternal alleles in induced pluripotent stem cells (iPSCs). Knockdown of SETDB1 in PWS-specific iPSCs causes a decrease in the accumulation of H3K9 trimethylation (H3K9me3) at SNORD116. We also show that upon knockdown of SETDB1 in PWS-specific iPSCs, expression of maternally silenced 116HG RNA is partially restored. SETDB1 knockdown in PWS iPSCs also disrupts DNA methylation at the PWS-IC where a decrease in 5-methylcytosine is observed in association with a concomitant increase in 5-hydroxymethylcytosine. In iPSCs-derived neurons and stem cells from human exfoliated teeth (SHEDs) ZNF274/SETDB1 complex binding and H3K9me3 modification occur in both alleles. These observations suggest that the ZNF274/SETDB1 complex bound to the SNORD116 cluster may protect the PWS-IC from DNA demethylation during early development, as indicated by iPSCs. Our findings reveal novel epigenetic mechanisms that function to repress the maternal 15q11-q13 region. The better understanding of epigenetic mechanisms provides new tools for future therapy research.
4

Eddiry, Sanaa. "Rôle du SNORD116 et de l'IGFBP7 dans la réponse à l'IGF1 dans le syndrome de Prader-Willi." Toulouse 3, 2013. http://www.theses.fr/2013TOU30215.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Le syndrome de Prader-Willi (SPW) est une maladie génétique complexe du développement qui résulte de l'absence d'expression de gènes sur le chromosome15q11-q13 paternel. Les patients SPW présentent des taux de GH diminués et des taux de ghréline élevés. Ils sont traités précocement par GH. La région du chromosome 15q11-q13 responsable du SPW est soumise au phénomène d'empreinte génétique, des études récentes la limitent à une région minimale incluant un cluster de small nucleolar RNA (snoARN): le SNORD116. Nos résultats montrent une sensibilité accrue à l'IGF1 et à l'insuline dans les fibroblastes de patients SPW. Ces cellules montrent également un taux augmenté de la prolifération et une diminution de la sénescence. Des études par microarrays, RT-qPCR, ainsi que du sécrétome montrent que l'expression de l'IGFBP7, un important facteur anti-prolifératif, a été considérablement abaissée chez ces patients. IGFBP7 est connu pour interagir avec les récepteurs de l'IGF1 et de l'insuline en modulant négativement leur action. Ces résultats étaient identiques chez la patiente SD. Notre hypothèse a été que l'augmentation de la prolifération et de la sensibilité aux facteurs de croissance est due à l'absence de l'expression du SNORD116. Nous avons démontré que le défaut du SNORD116 entraîne des taux de prolifération élevés et une diminution de la sénescence chez les patients SPW, avec une diminution de la sécrétion de l'IGFBP7. Les taux d'IGFBP7 in vitro décroissent sous l'effet de l'IGF1. De plus nous avons constaté que l'augmentation des taux d'IGF1 était corrélée significativement avec la diminution des taux de l'IGFBP7 chez des enfants SPW traités par GH pendant un an. Ces études soulignent fortement l'importance du SNORD116 pour contrôler la production de l'IGFBP7 en présence d'IGF1 et de facteurs de croissance et donc la sensibilité au traitement à l'hormone de croissance
Prader-willi syndrome (PWS) is a complex genetic disease of neurodevelopment that arises from lack of expression of paternally imprinted genes on chromosome 15q11-q13. GH levels are low in PWS, and GH treatment is recommended. The current management of PWS patients includes early treatment by growth hormone (GH). We demonstrated that GH treatment of PWS patients is associated with elevated IGF1 levels. Human chromosome 15q11-q13 contains an imprinting control region, which when deleted is sufficient to cause PWS. In addition, human genetic studies have defined a minimal PWS gene locus including a cluster of paternally expressed small nucleolar RNA (snoRNA), within the SNORD116. This makes PWS the first human disease found to be caused by loss of non-coding RNA. Our results showed increased sensitivity to IGF1 and Insulin in PWS cells. These cells demonstrate also increased proliferation rate and decreased senescence. From multi-array and RT-qPCR analysis, expression of IGFBP7, an important antiproliferative factor, was dramatically decreased in those patients. IGFBP7 is known to interact with IGF1 and Insulin receptors to decrease their action. We demonstrated that the lack of expression of SNORD116 in this patient results in increased response to IGF1 and Insulin and highly decreased secretion of IGFBP7. Therefore lack of SNORD116 results in high proliferation rate and decreased senescence in PWS, with decreased IGFBP7 secretion. Finally, we found that the increase of IGF1 level was significantly correlated with the decrease of IGFBP7 level in the serum of PWS children treated one year with GH. These data suggest that the lack of SNORD116 expression results in increased responsiveness to growth factors due to a low level of IGFBP7 in cells of PWS patients. They highlight a new phenotype of PWS, modified IGFBP7 levels, which, given the properties of IGFBP7 as a strong regulator of IGF1 effect, has potential consequences on the management of PWS patients treated by GH
5

Cole, Lisa. "The Role of SNORD116 in the Neuromolecular Pathogenesis of the Prader-Willi Syndrome." Thesis, 2016. https://doi.org/10.7916/D8XW4K0X.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Prader-Willi syndrome (PWS) is caused by a loss of paternally expressed genes in an imprinted region of chromosome 15q. Among the canonical PWS phenotypes are hyperphagic obesity, central hypogonadism, and low growth hormone. Rare microdeletion PWS patients define a 91 kb minimum critical deletion region encompassing three genes, including the non-coding SNORD116. Induced pluripotent stem cells were generated from skin cells of three large deletion (5-6 Mb) PWS patients and one novel microdeletion (118 kb) PWS patient. We found that genes within the PWS region, including SNRPN and NDN, showed persistence of DNA methylation after iPSC reprogramming and differentiation to neurons. Genes within the PWS minimum critical deletion region remain silenced in both PWS large deletion and microdeletion iPSC following reprogramming. We find that NHLH2 and PC1 (protein and transcript) are reduced in PWS patient iPSC-derived neurons. Nhlh2 and Pcsk1 expression are reduced in hypothalami of fasted Snord116p-/m+ mice while hypothalamic AgRP and Npy remain elevated following refeeding in association with relative hyperphagia. Nhlh2-/- mice have growth deficiencies from 4-7 weeks of age, develop hyperphagic obesity as adults, and are hypogonadal. Nhlh2 promotes expression of the prohormone convertase, Pcsk1 (PC1). PC1 is a neuroendocrine prohormone convertase that catalyzes the processing of hormones to “mature,” active hormones. Humans and mice deficient in PC1 display hyperphagic obesity, hypogonadism, decreased growth hormone, and hypoinsulinemic diabetes due to impaired prohormone processing. Snord116p-/m+ mice display in vivo functional defects in prohormone processing of proinsulin, proGHRH, and proghrelin in association with reductions in islet, hypothalamic, and stomach PC1 content. Our findings suggest that the major neuroendocrine features of PWS are due to PC1 deficiency which results from absence of functional SNORD116. In addition to hyperphagic obesity and endocrinopathies, global developmental delay (delayed motor milestones, delayed language development) is a major characteristic of the Prader-Willi syndrome (PWS). We identified neuroanatomical defects in iPSC-derived neurons of individuals with PWS and mice deficient for Snord116. iPSC-derived neurons from PWS patients and neurons from Snord116p-/m+ mice, have smaller soma and decreased numbers of neurites. Reduced neuron cell body size is apparent in utero and persists at least until 4 weeks of age in Snord116p-/m+ mice. The reduction in neuronal soma size is associated with smaller neuronal nucleoli. There are also developmental defects in the endocrine pancreas of Snord116p-/m+ animals that persist into adulthood (≥20 weeks). Mice lacking Snord116 have smaller pancreatic islets and within the islet the percentage of δ-cells is increased, while the percentage of α-cells is reduced. In Snord116p-/m+ isolated islets, Sst and Hhex are upregulated while Ins1, Ins2, Pdx1, Nkx6-1, and Pax6 are downregulated. There is a 3-fold increase in the percentage of polyhormonal cells in the neonatal islets of Snord116p-/m+ mice, which was due to an increase in cells co-positive with somatostatin. Snord116 may play a role in islet cell lineage specification. Overall, this work suggests that the Snord116 gene cluster is important for developmental processes in the brain as well as endocrine pancreas and prohormone processing in multiple tissues. Loss of elements within this cluster could account for the PWS by virtue of effects on the expression of PCSK1.

Частини книг з теми "SNORD115":

1

Duis, Jessica. "Behavioral Complications Associated With Snord116 Deletion in Prader-Willi Syndrome." In Encyclopedia of Behavioral Neuroscience, 2nd edition, 307–12. Elsevier, 2022. http://dx.doi.org/10.1016/b978-0-12-819641-0.00094-3.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.

До бібліографії