• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 11
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 16
  • 12
  • 12
  • 6
  • 5
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Role of the CHD7 chromatin remodeler protein in glioblastoma multiforme / Papel do remodelador de cromatina CHD7 em glioblastoma multiforme

Machado, Raquel Arminda Carvalho 15 June 2018 (has links)
Chromatin remodeler proteins exert an important function in promoting dynamic modifications in the chromatin architecture, rendering the transcriptional machinery available to the condensed genomic DNA. Due to this central role in regulating gene transcription, deregulation of these molecular machines may lead to severe perturbations in the normal cell functions. Loss-of-function mutations in the CHD7 gene, a member of the chromodomain helicase DNA-binding (CHD) family, are the major cause of the CHARGE syndrome in humans. The disease is characterized by a variety of congenital anomalies, including malformations of the craniofacial structures, peripheral nervous system, ears, eyes and heart. In this context, several studies have already shown the importance of CHD7 for proper function of the neural stem cells (NSCs). Interestingly, we found that CHD7 mRNA levels are upregulated in gliomas, when compared to normal brain tissue, therefore, we hypothesized that CHD7 might have a role in the pathogenesis of these tumors. To investigate the possible oncogenic role of CHD7 in glioblastoma (GBM), we adopted gain- and loss-of-function approaches in adherent GBM cell lines. Using CRISPR_Cas9 genome editing, we found that CHD7 deletion suppresses anchorage-independent growth and reduces spheroid invasion in human LN-229 cells. Moreover, deletion of CHD7 delayed tumor growth and improved overall survival in an orthotopic xenograft glioma mouse model. Conversely, ectopic overexpression of CHD7 in LN-428 and A172 cells was found to increase cell motility and invasiveness in vitro and LN-428 tumor growth in vivo. RNAseq analysis showed that alterations of CHD7 expression levels promote changes in several molecular pathways and modulate critical genes associated with cell adhesion and locomotion. However, the mechanisms underlying the effects of CHD7 overexpression in glioma tissue are still not understood. Here, we also generated recombinant plasmid with functional CHD7 promoter activity reported by luciferase assay. This powerful tool should enable future studies to determine the direct targeting relationship between different signal transduction pathways and CHD7 geneexpression. In summary, our findings indicate that GBM cells expressing a high level of CHD7 may exist and contribute to tumor infiltration and recurrence. Further studies should warrant important clinical-translational implications of our findings for GBM treatment. / As proteínas remodeladoras de cromatina exercem importante papel, promovendo modificações dinâmicas na arquitetura da cromatina e dando acesso à maquinaria transcricional ao DNA genômico condensado. Devido à esta função central na regulação da transcrição gênica, a desregulação dessas máquinas moleculares pode levar a perturbações graves na função normal das células. Assim, por exemplo, mutações do tipo perda de função no gene CHD7, um membro da família \"chromodomain helicase DNA-binding\" (CHD), são a principal causa da síndrome de CHARGE em humanos. A doença é caracterizada por uma variedade de anomalias congênitas, incluindo malformações das estruturas craniofaciais, sistema nervoso periférico, orelhas, olhos e coração. Neste contexto, vários estudos já mostraram a importância da proteína CHD7 para o funcionamento normal de células-tronco neurais (NSCs). Curiosamente, descobrimos que os níveis de mRNA de CHD7 estão mais fortemente expressos em gliomas, quando comparados ao tecido cerebral normal, portanto, nós hipotetizamos que CHD7 poderia ter um papel na patogênese desses tumores. Para investigar o possível papel oncogênico de CHD7 em glioblastoma (GBM), utilizamos enfoques de ganho e perda de função em linhagens celulares aderentes de GBM. Utilizando a técnica de CRISPR_Cas9 para edição do genoma, demonstramos que a deleção do gene CHD7 suprime o crescimento independente de ancoragem e reduz a invasão de esferóides em células LN-229 humanas de GBM. Além disso, a deleção de CHD7 reduziu o crescimento do tumor e melhorou a sobrevida em modelo de injeção ortotópica xenográfica em camundongo. Por outro lado, verificou-se que a super-expressão ectópica de CHD7 nas células LN-428 e A172 aumenta não só a motilidade celular e a capacidade de invasão in vitro, mas, também, o crescimento do tumor de LN-428 in vivo. A análise de RNA-seq mostrou que o nocauteamento da sequência codificadora de CHD7 e sua super-expressão promovem alterações em diversas vias moleculares, modulando genes críticosassociados à adesão e locomoção celular. No entanto, os mecanismos subjacentes aos efeitos da super-expressão de CHD7 em tecidos de glioma ainda não são compreendidos. Neste trabalho, geramos um plasmídeo recombinante contendo um fragmento da região promotora de CHD7, o qual se mostrou funcional em ensaios de luciferase. Esta ferramenta permitirá que estudos futuros possam identificar a relação direta entre as diferentes vias de transdução de sinal e a expressão do gene CHD7. Em resumo, nossos achados indicam que células de GBM expressando um alto nível de CHD7 podem existir e contribuir para a infiltração e recorrência do tumor. Estudos posteriores deverão avaliar as possíveis implicações dos resultados apresentados neste trabalho para a translação clínica no tratamento de pacientes com GBM.
2

Role of the CHD7 chromatin remodeler protein in glioblastoma multiforme / Papel do remodelador de cromatina CHD7 em glioblastoma multiforme

Raquel Arminda Carvalho Machado 15 June 2018 (has links)
Chromatin remodeler proteins exert an important function in promoting dynamic modifications in the chromatin architecture, rendering the transcriptional machinery available to the condensed genomic DNA. Due to this central role in regulating gene transcription, deregulation of these molecular machines may lead to severe perturbations in the normal cell functions. Loss-of-function mutations in the CHD7 gene, a member of the chromodomain helicase DNA-binding (CHD) family, are the major cause of the CHARGE syndrome in humans. The disease is characterized by a variety of congenital anomalies, including malformations of the craniofacial structures, peripheral nervous system, ears, eyes and heart. In this context, several studies have already shown the importance of CHD7 for proper function of the neural stem cells (NSCs). Interestingly, we found that CHD7 mRNA levels are upregulated in gliomas, when compared to normal brain tissue, therefore, we hypothesized that CHD7 might have a role in the pathogenesis of these tumors. To investigate the possible oncogenic role of CHD7 in glioblastoma (GBM), we adopted gain- and loss-of-function approaches in adherent GBM cell lines. Using CRISPR_Cas9 genome editing, we found that CHD7 deletion suppresses anchorage-independent growth and reduces spheroid invasion in human LN-229 cells. Moreover, deletion of CHD7 delayed tumor growth and improved overall survival in an orthotopic xenograft glioma mouse model. Conversely, ectopic overexpression of CHD7 in LN-428 and A172 cells was found to increase cell motility and invasiveness in vitro and LN-428 tumor growth in vivo. RNAseq analysis showed that alterations of CHD7 expression levels promote changes in several molecular pathways and modulate critical genes associated with cell adhesion and locomotion. However, the mechanisms underlying the effects of CHD7 overexpression in glioma tissue are still not understood. Here, we also generated recombinant plasmid with functional CHD7 promoter activity reported by luciferase assay. This powerful tool should enable future studies to determine the direct targeting relationship between different signal transduction pathways and CHD7 geneexpression. In summary, our findings indicate that GBM cells expressing a high level of CHD7 may exist and contribute to tumor infiltration and recurrence. Further studies should warrant important clinical-translational implications of our findings for GBM treatment. / As proteínas remodeladoras de cromatina exercem importante papel, promovendo modificações dinâmicas na arquitetura da cromatina e dando acesso à maquinaria transcricional ao DNA genômico condensado. Devido à esta função central na regulação da transcrição gênica, a desregulação dessas máquinas moleculares pode levar a perturbações graves na função normal das células. Assim, por exemplo, mutações do tipo perda de função no gene CHD7, um membro da família \"chromodomain helicase DNA-binding\" (CHD), são a principal causa da síndrome de CHARGE em humanos. A doença é caracterizada por uma variedade de anomalias congênitas, incluindo malformações das estruturas craniofaciais, sistema nervoso periférico, orelhas, olhos e coração. Neste contexto, vários estudos já mostraram a importância da proteína CHD7 para o funcionamento normal de células-tronco neurais (NSCs). Curiosamente, descobrimos que os níveis de mRNA de CHD7 estão mais fortemente expressos em gliomas, quando comparados ao tecido cerebral normal, portanto, nós hipotetizamos que CHD7 poderia ter um papel na patogênese desses tumores. Para investigar o possível papel oncogênico de CHD7 em glioblastoma (GBM), utilizamos enfoques de ganho e perda de função em linhagens celulares aderentes de GBM. Utilizando a técnica de CRISPR_Cas9 para edição do genoma, demonstramos que a deleção do gene CHD7 suprime o crescimento independente de ancoragem e reduz a invasão de esferóides em células LN-229 humanas de GBM. Além disso, a deleção de CHD7 reduziu o crescimento do tumor e melhorou a sobrevida em modelo de injeção ortotópica xenográfica em camundongo. Por outro lado, verificou-se que a super-expressão ectópica de CHD7 nas células LN-428 e A172 aumenta não só a motilidade celular e a capacidade de invasão in vitro, mas, também, o crescimento do tumor de LN-428 in vivo. A análise de RNA-seq mostrou que o nocauteamento da sequência codificadora de CHD7 e sua super-expressão promovem alterações em diversas vias moleculares, modulando genes críticosassociados à adesão e locomoção celular. No entanto, os mecanismos subjacentes aos efeitos da super-expressão de CHD7 em tecidos de glioma ainda não são compreendidos. Neste trabalho, geramos um plasmídeo recombinante contendo um fragmento da região promotora de CHD7, o qual se mostrou funcional em ensaios de luciferase. Esta ferramenta permitirá que estudos futuros possam identificar a relação direta entre as diferentes vias de transdução de sinal e a expressão do gene CHD7. Em resumo, nossos achados indicam que células de GBM expressando um alto nível de CHD7 podem existir e contribuir para a infiltração e recorrência do tumor. Estudos posteriores deverão avaliar as possíveis implicações dos resultados apresentados neste trabalho para a translação clínica no tratamento de pacientes com GBM.
3

The role of Chd7 & Chd8 chromatin remodelers in oligodendrogenesis and (re)myelination / Le rôle de Chd7 & Chd8 facteurs du remodelage de la chromatine dans l'oligodendrogenese et la (re)myélinisation

Marie, Corentine 29 September 2017 (has links)
Les oligodendrocytes (OLs) sont les cellules myélinisantes du système nerveux central, s’enroulant autour des axones et permettant la conduction saltatoire du potentiel d’action. Dans la Sclérose en Plaques, des gaines de myélines sont détruites et l’efficacité de la remyélinisation par les précurseurs d’oligodendrocytes (OPCs) diminue avec la progression de la maladie. Une meilleure compréhension du mécanisme qui contrôle la génération des OPCs et leur différentiation est donc essentielle pour développer des thérapies efficaces de remyélinisation. L’oligodendrogenèse, qui comprend les étapes de génération des OPCs, de différenciation et de maturation des OLs, est un processus contrôlé par des facteurs de transcription spécifiques incluant Ascl1, Olig2 and Sox10 mais le mécanisme impliqué est encore peu connu. Sachant que les facteurs du remodelage de la chromatine sont des régulateurs nécessaires à la formation de la boucle promoter-enhancer permettant l’initiation de la transcription, nous nous sommes focalisé sur Chd7 (Chromodomain-Helicase-DNA-Binding 7), un membre de la famille de protéine CHD. Dans une première étude, nous avons montré que Chd7 est hautement enrichi dans le lignage oligodendroglial avec un pic d’expression pendant la différenciation des OLs. Nous avons également montré que la délétion conditionnelle de Chd7 diminuait la différentiation des OLs pendant la (re)myélinisation. Dans un seconde étude, nous avons utilisé des techniques de génomique sur les OPCs purifiés pour étudier la régulation par Chd7 de gènes impliqués dans la différenciation, la survie et la prolifération des OPCs. Dans ce but, nous avons générer des délétions inductible de Chd7 spécifiquement dans les OPCs (Chd7iKO) et nous avons analysé le transcriptome (RNA-seq) d’OPCs purifiés à partir de cerveaux de souris P7 comparé à des contrôles. Nous avons trouvé que Chd7 activait l’expression des gènes impliqué dans la différenciation des OPCs et la myélinisation et inhibait l’apoptose, sans montré de défaut de prolifération. Pour aller plus loin, nous avons étudié Chd8, un paralogue de Chd7, et nous avons montré qu’il est exprimé dans le lignage oligodendrocytaire avec un pic d’expression dans les OL en différenciation, similairement à Chd7. Les données de fixation (ChIP-seq) de Chd7 et Chd8 indiquent que ces deux facteurs du remodelage de la chromatine se fixent sur des gènes communs reliés au processus de différenciation, de survie et de prolifération des OPCs. Intégrant ces données avec celles de facteurs transcriptionnels clés dans l’oligodendrogenèse (Olig2, Ascl1 et Sox10), nous avons construit un modèle de la régulation de l’expression de gènes contrôlés dans le temps et impliqué dans chacune des étapes de la différenciation des oligodendrocytes. / Oligodendrocytes (OLs) are myelin-forming cells of the central nervous system wrapping axons and allowing the saltatory conduction of action potentials. In Multiple sclerosis (MS), myelin sheath is destroyed and effective remyelination by oligodendrocyte precursor cells (OPCs) diminishes with disease progression. Therefore, a better understanding of the mechanisms controlling OPC generation and differentiation is essential to develop efficient remyelinating therapies. Oligodendrogenesis, involving the steps of OPC generation, OPC differentiation and maturation of OLs, is a process controlled by specific transcription factors including Ascl1, Olig2 and Sox10 but the mechanisms involved are poorly understood. As it is known that chromatin remodelers are regulatory factors necessary in the formation of the promoter-enhancer loop prior to transcription, we focused our study on Chd7 (Chromodomain-Helicase-DNA-Binding 7), a member of the CHD protein family. In a first study, we showed that Chd7 is highly enriched in the oligodendroglial lineage cells with a peak of expression during OL differentiation and that Chd7 OPC-conditional deletion impairs OL differentiation during (re)myelination. In a second study, we used unbiased genome wide technics in purified OPCs to study Chd7 regulation of genes involved in OPC differentiation, proliferation and survival. To this aim, we have generated OPC-specific inducible Chd7 knock-out (Chd7-iKO) and analyse the transcriptome (RNA-seq) of purified OPCs from P7 mouse cortices compared to control littermates. We found that Chd7 promote the expression genes involved in OPC differentiation and myelination and inhibits apoptosis, without affecting OPC proliferation. Furthermore, we investigated Chd8, a paralog of Chd7, showing that it is expressed in the oligodendroglial lineage with a peak of expression in differentiating oligodendrocytes, similar to Chd7. Genome wide binding (ChIP-seq) profiling for Chd7 and Chd8 indicate that these two chromatin remodelers bind to common genes related to OPC differentiation, survival and proliferation. Integrating these datasets with other key transcriptional regulators of oligodendrogenesis (Olig2, Ascl1 & Sox10), we have built a model accounting for the time-controlled regulate expression of genes involved in each step of OL differentiation.
4

Epigenetic Regulation of Hematopoiesis in Zebrafish

Huang, Hsuan-Ting 02 November 2012 (has links)
The initiation of the hematopoietic program is orchestrated by key transcription factors that recruit chromatin regulators in order to activate or inhibit blood target gene expression. To generate a complete compendium of chromatin factors that establish the genetic code during developmental hematopoiesis, we conducted a large-scale reverse genetic screen targeting 425 chromatin factors in zebrafish and identified over 30 novel chromatin regulators that function at distinct steps of embryonic hematopoiesis. In vertebrates, developmental hematopoiesis occurs in two waves. During the first and primitive wave, mainly erythrocytes are produced, and we identified at least 15 chromatin factors that decrease or increase formation of \(scl^+\), \(gata1^+\), and \(\beta-globin e3^+\) erythroid progenitors. In the definitive wave, HSCs capable of self-renewal and differentiation into multiple lineages are induced, and we identified at least 18 chromatin factors that decrease or increase the formation of \(c-myb^+\) and \(runx1^+\) stem and progenitor cells in the aorta gonad mesonephros (AGM) region, without disruption of vascular development. The majority of the chromatin factors identified from the screen are involved in histone acetylation, histone methylation, and nucleosome remodeling, the same modifications that are hypothesized to have the most functional impact on the transcriptional status of a gene. Moreover, these factors can be mapped to subunits of chromatin complexes that modify these marks, such as HBO/HAT, HDAC/NuRD, SET1A/MLL, ISWI, and SWI/SNF. One of the strongest phenotypes identified from the screen came from knockdown of chromodomain helicase DNA binding domain 7 (chd7). Morpholino knockdown of chd7 resulted in increased primitive and definitive blood production from the induction of stem and progenitor cells to the differentiation of myeloid and erythroid lineages. This expansion of the blood lineage is cell autonomous as determined by blastula transplantation experiments. Though chromatin factors are believed to function broadly and are often expressed ubiquitously, the combined results of the screen and chd7 analysis show that individual factors have very tissue specific functions. These studies implicate chromatin factors as playing a major role in establishing the programs of gene expression for self-renewal and differentiation of hematopoietic cells.
5

Investigation of the Molecular Function of CHD7, the Protein Implicated in CHARGE Syndrome, Using Next-Generation Genomics

Schnetz, Michael Paul January 2010 (has links)
No description available.
6

Investigation of CHD7 Function in Developmental Models of CHARGE Syndrome

Balow, Stephanie Ann 11 June 2014 (has links)
No description available.
7

Development of a functional assay for CHD7, a protein involved in CHARGE syndrome / Mise au point d'un test fonctionnel pour la protéine CHD7 impliquée dans le syndrome CHARGE

Brajadenta, Gara Samara 14 June 2019 (has links)
Le syndrome CHARGE (CS) est une maladie génétique rare caractérisée par de nombreuses anomalies congénitales, majoritairement causées par des altérations de novo du gène CHD7. Celui-ci code pour une protéine à chromodomaines, impliquée dans le remodelage ATP-dépendant de la chromatine. La grande majorité des altérations de CHD7 consiste en allèles nuls tels que des délétions, des substitutions non-sens ou des décalages du cadre de lecture. Nous avons réalisé le premier diagnostic moléculaire d’un patient Indonésien atteint du CS, en étudiant un panel de gènes (CHD7, EFTUD2, et HOXA1) par NGS (next-generation sequencing). Nous avons identifié une nouvelle mutation non-sens hétérozygote dans l’exon 34 du gène CHD7 (c.7234G>T ou p.Glu2412Ter). Par ailleurs, il n'existe pas d’analyse fonctionnelle qui permettrait de caractériser la pathogénicité des variants de la protéine CHD7 rencontrés chez des patients. C’est pourquoi l’objectif de ce travail est de mettre au point un test fonctionnel de la protéine CHD7, sous forme sauvage ou mutée. Pour cela, nous avons généré par mutagénèse dirigée des vecteurs codant pour trois variants faux-sens de CHD7 et le variant présentant une insertion de cinq acides aminés. Ensuite, les protéines CHD7, sous forme sauvage ou variante, ont été surexprimées dans la lignée HeLa. L’expression des protéines a été mise en évidence par western blot et par immunofluorescence. Pour étudier la fonctionnalité de CHD7, nous avons quantifié par RT-qPCR les transcrits de cinq gènes (l’ADNr 45S, SOX4, SOX10, MYRF, et ID2), dont la transcription est selon le littérature régulée par CHD7. Nous avons observé que l’expression de CHD7 sauvage entraînait une diminution significative et reproductible des quantités de transcrits correspondant à tous les gènes rapporteurs. Par contre, l’expression des quatre allèles variants de CHD7 n’avait aucun impact, ce qui suggère que ces variants ne sont pas fonctionnels. Par ailleurs, nous avons appliqué notre test biologique dans des cellules de la lignée SH-SY5Y, pour lesquelles nous avons introduit une mutation faux-sens dans le génome en utilisant la technique CRISPR/Cas9. Lorsque ce variant était exprimé, les niveaux de transcription des cinq gènes rapporteurs n’étaient pas significativement différents de ceux observés dans les cellules où les deux allèles de CHD7 avaient été invalidés. Par conséquent, les variants étudiés peuvent être répertoriés comme résultant de mutations causales du CS. / CHARGE syndrome (CS) is a rare genetic disease characterized by numerous congenital abnormalities, mainly caused by de novo alterations of the CHD7 gene. It encodes a chromodomain protein, involved in the ATP-dependent remodeling of chromatin. The vast majority of CHD7 alterations consists in null alleles like deletions, non-sense substitutions or frameshift-causing variations. We report the first molecular diagnosis of an Indonesian CS patient by a targeted NGS (next-generation sequencing) gene panel (CHD7, EFTUD2, and HOXA1). We identified a novel heterozygous nonsense mutation in exon 34 of CHD7 (c.7234G>T or p.Glu2412Ter). Functional analyses to confirm the pathogenicity of CHD7 variants are lacking and urgently needed. Therefore, the aim of this study was to establish a functional test for wild-type (WT) or variants of CHD7 protein found in CS patients. Using an expression vector encoding CHD7, three variants harboring an amino acid substitution and one variant with a five-amino acid insertion were generated via site-directed mutagenesis. Then CHD7 proteins, either wild-type (WT) or variants, were overexpressed in HeLa cell line. Protein expression was highlighted by western blot and immunofluorescence. We then used real-time RT-PCR to study CHD7 functionality by evaluating the transcript amounts of five genes whose expression is regulated by CHD7 according to the literature. These reporter genes are 45S rDNA, SOX4, SOX10, ID2, and MYRF. We observed that, upon WT-CHD7 expression, the reporter gene transcriptions were downregulated, whereas the four variant alleles of CHD7 had no impact. This suggests that these alleles are not polymorphisms because the variant proteins appeared non-functional. Furthermore, we applied our biological assay in SH-SY5Y cell line in which endogenous CHD7 gene was mutated using the CRISPR/Cas9 technique. Then, we observed that when a CHD7 missense variant was expressed, the transcription levels of the five reporter genes were non-significantly different, compared with the cells in which both CHD7 alleles were knocked-out. Therefore, the studied variants can be considered as disease-causing of CS.
8

Le syndrome CHARGE : étude clinique et moléculaire / Clinical and molecular aspects of CHARGE syndrome

Legendre, Marine 14 December 2016 (has links)
Le syndrome CHARGE est une association malformative rare due à une mutation du gène CHD7 dans 60 à 90% des cas. L'objectif de ce travail était d'en décrire les éléments cliniques et moléculaires afin d'optimiser la prise en charge de patients atteints d'un handicap multisensoriel lourd.Le diagnostic anténatal en est difficile et l'étude de 40 fœtus a permis d'affiner la description du phénotype, de décrire de nouveaux éléments cliniques et finalement de proposer un ajustement des critères diagnostiques chez le fœtus.L'étude endocrinienne de 42 patients confirme la présence d'un hypogonadisme hypogonadotrope dans 97% des cas. Souvent méconnu et non traité il peut avoir des conséquences délétères sur la qualité de vie. Nous proposons qu'il soit reconnu comme critère majeur du syndrome.L'étude clinique d'une cohorte française de 119 patients a montré que la surdité et l'atteinte des canaux semi circulaires sont les éléments les plus fréquents du syndrome (suivis de l'atteinte hypophysaire, de l'arhinencéphalie et des anomalies de l'oreille externe), et les seuls significativement associés à la présence d'une mutation dans CHD7. Une étude approfondie des données issues de cette étude est en cours.Sur le plan moléculaire, alors que la plupart des mutations identifiées sont des mutations tronquantes privées apparues de novo, nous avons identifié 4 variants au niveau de l'intron 25, récurrents pour certains, dont l'interprétation était délicate. Leur étude in silico puis par une technique de minigène a permis de mettre en évidence une configuration particulière des séquences impliquées dans l'épissage de l'exon 26 (point de branchement distant) et de démontrer leur pathogénicité. / CHARGE syndrome is a rare disorder of multiple congenital anomalies ascribed to a CHD7 gene mutation in 60% to 90% of cases. The aim of this study was to improve the knowledge regarding molecular and clinical aspects of the syndrome in order to optimize the management of these patients with severe disability. Antenatal diagnosis remains challenging in many instances and a detailed clinicopathological survey in a series of 40 fetuses allowed us to refine the clinical description of CHARGE syndrome in fetuses, describe some novel features and set up diagnostic criteria. An endocrinologic study of 42 patients showed a hypogonadotrophic hypogonadism in 97% of cases. For this reason, it should be considered as a major symptom of the syndrome. An early screening should lead to a hormonal replacement therapy which dramatically impacts the condition.A study of a French cohort of 119 patients found that deafness and semi-circular canals hypoplasia were the most frequent symptoms (followed by hypogonadotrophic hypogonadism, arhinencephaly and external ears anomalies) and the only features statistically associated with a mutation in the CHD7 gene. A detailed study of the data is still going on.The syndrome is mainly due to de novo and private truncating mutations of the CHD7 gene but we report an intriguing hot spot of intronic mutations located in IVS25. Combining computational in silico analysis and ex vivo minigene assays, we explained this mutation hot spot by a particular genomic context, including a distant branch point, and confirmed the pathogenicity of these mutations.
9

Haplotypanalyse bei Familien mit einem Kind mit CHARGE- Syndrom und Kandidatengenscreening CHD8 und FAM124B / Haplotype analysis in families with CHARGE syndrome affected child and screening of candidate genes CHD8 and FAM124B

von Velsen, Nina 31 July 2018 (has links)
No description available.
10

CHARGE syndrome: candidate genes and pathogenesis

Schulz, Yvonne 14 October 2014 (has links)
No description available.

Page generated in 0.4099 seconds