Sommaire
Littérature scientifique sur le sujet « Réparation des mésappariement »
Créez une référence correcte selon les styles APA, MLA, Chicago, Harvard et plusieurs autres
Consultez les listes thématiques d’articles de revues, de livres, de thèses, de rapports de conférences et d’autres sources académiques sur le sujet « Réparation des mésappariement ».
À côté de chaque source dans la liste de références il y a un bouton « Ajouter à la bibliographie ». Cliquez sur ce bouton, et nous générerons automatiquement la référence bibliographique pour la source choisie selon votre style de citation préféré : APA, MLA, Harvard, Vancouver, Chicago, etc.
Vous pouvez aussi télécharger le texte intégral de la publication scolaire au format pdf et consulter son résumé en ligne lorsque ces informations sont inclues dans les métadonnées.
Articles de revues sur le sujet "Réparation des mésappariement"
Brunori, Michèle, Éric Gilson et Alain Puisieux. « Le système de réparation des mésappariements contrôle la stabilité des télomères ». médecine/sciences 17, no 11 (novembre 2001) : 1184–86. http://dx.doi.org/10.1051/medsci/200117111184.
Texte intégralOlschwang, S., et F. Eisinger. « Prédisposition héréditaire au cancer colorectal et inactivation de la fonction de réparation des mésappariements de l'ADN ». EMC - Gastro-entérologie 1, no 1 (janvier 2006) : 1–6. http://dx.doi.org/10.1016/s1155-1968(05)38974-7.
Texte intégralOlschwang, S., et F. Eisinger. « Prédisposition héréditaire au cancer colorectal et inactivation de la fonction de réparation des mésappariements de l'ADN ». EMC - Hépato-Gastroenterologie 2, no 3 (juillet 2005) : 214–22. http://dx.doi.org/10.1016/j.emchg.2005.03.002.
Texte intégralOlschwang, S., et P. Grandval. « Prédisposition héréditaire au cancer colorectal et inactivation de la fonction de réparation des mésappariements de l’ADN ». EMC - Gastro-entérologie 40, no 3 (juillet 2023) : 1–10. https://doi.org/10.1016/s1155-1968(23)62910-4.
Texte intégralFrouin, E., C. Alleyrat, J. Godet, L. Karayan-Tapon, H. Sinson, F. Morel, J. C. Lecron et L. Favot. « Le microenvironnement immun des tumeurs sébacées dépend de l’agressivité des tumeurs mais pas des altérations de la voie de réparation des mésappariements ». Annales de Dermatologie et de Vénéréologie - FMC 4, no 8 (décembre 2024) : A88—A89. http://dx.doi.org/10.1016/j.fander.2024.09.519.
Texte intégralLopez, C., A. Al Ghuzlan, J. Hadoux, R. Libé, L. Lamartina, F. Pani, D. Déandreis et al. « Impact d’une mutation sur l’un des gènes du système de réparation des mésappariements (MMR) sur le phénotype clinique et la réponse aux traitements des corticosurrénalomes métastatiques ». Annales d'Endocrinologie 85, no 5 (octobre 2024) : 396. http://dx.doi.org/10.1016/j.ando.2024.08.131.
Texte intégralAgiannidis, Christos, Zoe D. Pana, Dimitra Molyva, Konstantinos Kalokasidis et Maria Mixiou. « Metachronous Occurrence of Colorectal Cancer in a Muir-Torre Syndrome Patient Presenting with Recurrent Sebaceous Adenoma of the Eyelid : Case Report and Updated Review of the Literature ». Journal of Cutaneous Medicine and Surgery 16, no 6 (novembre 2012) : 394–99. http://dx.doi.org/10.1177/120347541201600607.
Texte intégralThèses sur le sujet "Réparation des mésappariement"
Hernandez-Pigeon, Hélène. « Caractérisation de nouveaux mécanismes de régulation des protéines de la réparation des mésappariements de l'ADN ». Toulouse 3, 2004. http://www.theses.fr/2004TOU30191.
Texte intégralSangar, Fatiha. « Étude fonctionnelle de SMAP1 : un nouveau gène à la croisée du trafic vésiculaire et de l’oncogenèse ». Paris 6, 2012. http://www.theses.fr/2012PA066121.
Texte intégralTumours that are deficient in mismatch repair system accumulate numerous mutations throughout the genome in repeated sequences, called microsatellites. Genes that contain microsatellite in their sequence are target genes of microsatellite instability (MSI). The aim of my thesis was to determine the functional consequences of mutations occurring in a newly identified target gene of the MSI oncogenesis : SMAP1 (Small ArfGAP1), which encodes a protein of the ArfGAP (ADP ribosylation factor GTPase Activating Protein) family specific for Arf6, a protein involved in clathrin-mediated endocytosis and actin remodelling. SMAP1 mutations are specific for MSI tumours of various tissue origins and occur in its coding poly (A)10 microsatellite. SMAP1 mutations are especially frequent in MSI colorectal cancers (CRC), in which its mutation frequency decreases with tumour progression suggesting that tumours devoid of SMAP1 mutations are more invasive. Functional experiments allowed us to show that SMAP1 mutations lead to a defect in the fast recycling step of the transferrin receptor trafficking, to increased cell proliferation and to decreased invasiveness by maintaining the adherent junctions. Interestingly, our observations showing that SMAP1 mutations increase the proliferative potential but reduce the invasiveness of MSI CRC-derived cell lines may explain some of the MSI CRCs clinical features, notably their large size and low metastatic potential
Monnet, Jordan. « Single-molecule analysis of the mismatch repair initiation and excision steps in Escherichia coli from a mechanical and kinetic standpoint= ». Paris 7, 2014. http://www.theses.fr/2014PA077202.
Texte intégralPostreplicative DNA mismatch repair is present in almost all organisme. Absence or failure of this system leads to mutator phenotype and, in humans, predisposition to cancer. In E. Coli the process is initiated by MutS that recognizes the mismatch and associates with an ATPase : MutL. Activation of the endonuclease MutH leads to nicking of the unmethylated single strand at the first hemymethylated 5'- GATC site (up to lkbp) close to the mismatch. Once the DNA is nicked, an helicase, UvrD, unwinds the DNA to extract and later excise the damaged strand to allow for its re-synthesis. In this PhD I propose a single-molecule analysis of the mismatch repair initiation and excision steps in Escherichia coli from a mechanical and kinetic standpoint. Using a single molecule technique, I have been able to show that a diffusion mechanism of proteins MutS and MutL from mismatch to a GATC site is the most likely, and to deciphe the mechanism by which UvrD is loaded at the nicking site to start the excision process
Borie, Claire. « Instabilité des microsatellites et cancers : recherche et description de ce phénotype tumoral dans les cancers du patient immunodéprimé ». Paris 7, 2011. http://www.theses.fr/2011PA077067.
Texte intégralMSI cancers arise as a consequence of loss of fonction of the DNA Mismatch repair system ("MMR" System). Inactivation of this system results in accumulation of replication errors in the genome of these tumors, in particular in repetitive séquences of the DNA called microsatellites; these tumors are therefore called MSI for " Instable Microsatellite". MSI cancers are inherited diseases associated with familial the colorectal cancer (Lynch syndrom or HNPCC), and represent 15-20 % of sporadic colorectal, gastric and endometrial cancers. Recently, the MSI phenotype was reported in other sporadic cancers. In particular owing to the works realized in our laboratory; this phenotype was described in non Hodgkin lymphomas (NHLs), but only in the NHL arising in an immuno-suppression context ( ID-RL), i. E. HIV-related or after organ transplantation. The objectives of my thesis were in this context: i) to specify the incidence of MSI in the ID-RL and establish the clinical and biological characteristics of these lymphomas compared with the other ID-RL (ID-RL not -MSI); ii) to search for a possible association between MSI and other frequent immuno-supression related tumoral types, e. G. In the squamous cell carcinomas cancers (ID-SCC) and the Kaposi's sarcomas (ID-KS). We established that the MSI phenotype is rarely observed in the ID-RL, representing 2-5 % of HIV-related NHL and approximately 10 % of PTLD. Briefly, MSI PTLD are characterized by their late occurrence after organ transplantation (median > 5 years), rare association with EBV (< 50 %), and frequent T phenotype (50 % of the PTLD T are MSI). MSI is significantly associated in these tumors after Azathioprine (Imurel) treatement. Further, loss of expression of the MMR proteins is heterogeneous in the ID-RL : MLH1, as well MSH2 or MSH6 can be involved. Loss of expression of O6 methyl guanine transferase (MGMT), a repair enzyme whose inactivation helps in selection of MMR deficient clones, was significantly associated with MSI in ID-RL. Finally, ID-RL MSI are characterized by few chromosomal rearangements and frequent mutations of the oncogene BRAF (Borie and al. , Int. J Cancer on 2010). My work involved the study of large series of tumors ID-RL, SCC and KS, to better understand the role of MSI in the oncogenetic process. Interestingly, I contributed to the identification of original clinical and molecular features of ID-RL MSI. Prospectives studies are needed to better define prognostic factors and therapeutic protocols of these tumors as compared with to the other ID-RL. This would be in particular interest, in PTLD-T. Following the example of what has been reported in other tumors, MSI could indeed be a factor to take into account in the treatment of these lymphomas
Laplante, Pierre. « Effect of MisMatch Repair Deficiency on metastasis occurrence modelized in a syngeneic mouse model ». Electronic Thesis or Diss., université Paris-Saclay, 2024. http://www.theses.fr/2024UPASL101.
Texte intégralBackground: It is established that mismatch repair deficient (MMR-D), hypermutated and microsatellite instable (MSI) colorectal cancer (CRC) is less prone to metastasis, and has worse prognosis after recurrence, than microsatellite stable (MSS) CRC. The mechanisms behind both these phenomena are still elusive, mainly due to the lack of a good in vivo model of metastatic MSI cancer.Methods: We generated Msh2 knockouts in the luciferase expressing breast cancer cell line 4T1luc+. The MMR-D phenotype, an accumulation of point mutations genome-wide and insertion-deletion (indels) at microsatellite sites, was established by serial passages in vitro before orthotopic injection in the lower left mammary fat pad of BALB/c mice. Metastasis development was assessed through in vivo imaging of luciferase activity. Primary tumors of mice bearing metastasis, or not, were sequenced, and genomic and transcriptomic analysis were performed. Spectral flow cytometry of CD45+ cells was carried out on primary tumors, secondary site (lung tissue), bone marrow and blood before and after tumor engraftment, to characterize the different microenvironments.Results: A significant difference in metastasis occurrence was noted between the MSS and MSI tumor-bearing mice (100% vs 82.3%, p = 0.0456). Moreover, MSI metastatic mice exhibited less site-specific metastatic development compared to their MSS counterparts. Whole exome sequencing (WXS) revealed that tumor mutation burden (TMB, non-synonymous mutations/megabase), indels, and MSIscore (percentage of instable microsatellites) were elevated in the MSI tumors compared to MSS, but did not explain the difference in metastasis occurrence. Transcriptomic analyses demonstrated that MSS tumors express epithelial-mesenchymal transition (EMT) related programs, while MSI metastatic and MSI localized primary tumor were enriched in proliferative and immune related signatures, respectively. Interestingly, MSI metastatic tumor were uniquely enriched with a hybrid EMT signature, a marker of cancer aggressiveness. Furthermore, spectral cytometry uncovered MSI-specific populations of tumor-associated macrophages and neutrophils (MSI-TAMs and MSI-TANs) at the primary tumor and metastatic sites. Finally, we discovered that MSI-TANs (but not MSI-TAMs) infiltration at secondary site predate metastatic development, and may participate in the creation of an MSI-specific pre-metastatic niche.Conclusion: We successfully developed a model of MSI metastatic cancer, recapitulating the hallmarks of the disease, namely elevated TMB, indels and MSIscore, as well as decreased metastatic potential. We have shown that metastatic development in our MSI model is associated with lower immune activity at the primary tumor, and that MSI metastatic tumors are enriched in proliferative and hybrid EMT signatures, that may explain the poor prognosis after recurrence in patients. At last, this model unveiled MSI-specific tumor-associated myeloid populations, and a potential MSI pre-metastatic niche. We provide here a model for fundamental and translational research on MSI metastatic disease, and provide insights on the crosstalk between metastasis development and the MSI phenotype
Dai, Jingqi. « Mécanisme moléculaire de l'endonucléase Mlh1-Mlh3 dans la voie de réparation des mésappariements de l’ADN et dans les processus de recombinaison en méiose ». Thesis, Université Paris-Saclay (ComUE), 2019. http://www.theses.fr/2019SACLS302.
Texte intégralMeiosis is key process in sexual reproduction, where chromosomes are segregated. During this process, a parental diploid cell divides into haploid gametes. This mechanism requires connections between homologous chromosomes and intermediate DNA structures called Holliday Junctions. These junctions are mainly resolved by MutLγ (Mlh1-Mlh3) complex. Mutations of genes involved in meiosis are associated with human diseases including sterility and chromosomal rearrangements such as trisomy. Mlh1-Mlh3 plays also a role in DNA mismatch repair (MMR). Our laboratory has characterized the first crystal structure of the C-terminal region of the MutLα complex (Mlh1-Pms1) which is the major endonuclease in MMR. My thesis aims at understanding the molecular mechanism of MutLγ (Mlh1-Mlh3) mainly involved in meiosis and to compare it with Mlh1-Pms1 mainly involved in MMR.We determined the crystal structure of the C-terminal domain of the MutLγ complex which contains the endonuclease site. We characterized the structure of three different states of the active site. We showed how Mlh1 is an integral part of the Mlh3 endonuclease site. We characterized the specificity of this domain for Holliday Junctions and proposed a model of the full-length complex and its DNA binding sites. Finally, we design new separation of function allele of Mlh1-Mlh3, called KERE, which was analyzed by EMSA and genetic experiments
Fan, Jun. « Single-molecule basis of transcription-coupled DNA repair ». Sorbonne Paris Cité, 2015. http://www.theses.fr/2015USPCC213.
Texte intégralThe DNA in living cells is constantly threatened by damages from both endogenous and exogenous agents, which can threaten genomic integrity, block processes of replication, transcription and translation and have also genotoxic effects. In response to the DNA damage challenge, organisms have evolved diverse surveillance mechanisms to coordinate DNA repair and cell-cycle progression. Multiple DNA repair mechanisms, discovered in both prokaryotic and eukaryotic organisms, bear the responsibility of maintaining genomic integrity; these mechanisms include nucleotide excision repair (NER), base excision repair (BER), mismatch repair (MMR) and double strand break repair (DSBR). Transcription-coupled DNA repair (TCR) is a specialized NER subpathway characterized by enhanced repair of the template strand of actively transcribed genes as compared to the classical global genome repair (GGR) subpathway of NER which does not distinguish between template and non-template strands. TCR achieves specialization via the involvement of RNA polymerase (RNAP) and the Mfd (Mutation Frequency Decline) protein, also known as TRCF (transcription repair coupling factor). TCR repair initiates when RNAP stalls at a DNA lesion on the transcribed strand and serves as the da mage sensor. The stalled RNAP must be displaced so as to make the lesion accessible to downstream repair components. E. Coli Mfd translocase participates in this process by displacing stalled RNAP from the lesion and then coordinating assembly of the UvrAB(C) components at th( damage site. Recent studies have shown that after binding to and displacing stalled RNAP, Mfd remains on the DNA in the form of a stable, translocating complex with evicted RNAP. So as to understand how UvrAB(C) are recruited via the Mfd-RNAP complex, magnetic trapping of individual, damaged DNA molecules was employed to observe-in real-time this multi¬component, multi-step reaction, up to and including the DNA incision reaction by UvrC. It was found that the recruitment of UvrA and UvrAB to the Mfd-RNAP complex halts the translocating complex and then causes dissolution of the complex in a molecular "hand-off" with slow kinetics Correlative single-molecule nanomanipulation and fluorescence further show that dissolution of the complex leads to loss of not only RNAP but also Mfd. Hand-off then allows for enhanced incision of damaged DNA by the UvrC component as compared to the equivalent single-moleculE GGR incision reaction. A global model integrating TCR and GGR components in repair was proposed, with the overall timescales for the parallel reactions provided
Aissi-Ben, Moussa Sana. « Caractérisation moléculaire des mutations germinales et somatiques associées au syndrome de Lynch en Tunisie ». Lille 2, 2009. http://www.theses.fr/2009LIL2S001.
Texte intégralHumbert, Odile. « Recombinaison entre séquences partiellement divergentes et réparation des mésappariements chez Streptococcus pneumoniae ». Toulouse 3, 1994. http://www.theses.fr/1994TOU30215.
Texte intégralTouat, Mahdi. « Mécanismes et implications thérapeutiques de l'hypermutation dans les gliomes Mechanisms and Therapeutic Implications of Hypermutation in Gliomas Mismatch Repair Deficiency in High-Grade Meningioma : A Rare but Recurrent Event Associated With Dramatic Immune Activation and Clinical Response to PD-1 Blockade Buparlisib in Patients With Recurrent Glioblastoma Harboring Phosphatidylinositol 3-Kinase Pathway Activation : An Open-Label, Multicenter, Multi-Arm, Phase II Trial Hyman DM. BRAF Inhibition in BRAFV600-Mutant Gliomas : Results From the VE-BASKET Study Glioblastoma Targeted Therapy : Updated Approaches From Recent Biology Successful Targeting of an ATG7-RAF1 Gene Fusion in Anaplastic Pleomorphic Xanthoastrocytoma With Leptomeningeal Dissemination Ivosidenib in IDH1-Mutated Advanced Glioma ». Thesis, université Paris-Saclay, 2020. http://www.theses.fr/2020UPASL071.
Texte intégralHigh tumor mutational burden (hypermutation) is observed in some gliomas; however, the mechanisms by which hypermutation develops and whether it predicts chemotherapy or immunotherapy response are poorly understood. Mechanistically, an association between hypermutation and mutations in the DNA mismatch-repair (MMR) genes has been reported in gliomas, but most MMR mutations observed in this context were not functionally characterized, and their role in causing hypermutation remains unclear. Furthermore, whether hypermutation enhances tumor immunogenicity and renders gliomas responsive to immune checkpoint blockade (e.g. PD-1 blockade) is not known. Here, we comprehensively analyze the clinical and molecular determinants of mutational burden and signatures in 10,294 gliomas, including 558 (5.4%) hypermutated tumors. We delineate two main pathways to hypermutation: a de novo pathway associated with constitutional defects in DNA polymerase and MMR genes, and a more common, post-treatment pathway, associated with acquired resistance driven by MMR defects in chemotherapy-sensitive gliomas recurring after temozolomide. Experimentally, the mutational signature of post-treatment hypermutated gliomas (COSMIC signature 11) was recapitulated by temozolomide-induced damage in MMR-deficient cells. While MMR deficiency was associated with acquired temozolomide resistance in glioma models, clinical and experimental evidence suggest that MMR-deficient cells retain sensitivity to the chloroethylating nitrosourea lomustine. MMR-deficient gliomas exhibited unique features including the lack of prominent T-cell infiltrates, extensive intratumoral heterogeneity, poor survival and low response rate to PD-1 blockade. Moreover, while microsatellite instability in MMR-deficient gliomas was not detected by bulk analyses, single-cell whole-genome sequencing of post-treatment hypermutated glioma cells demonstrated microsatellite mutations. Collectively, these results support a model where differences in the mutation landscape and antigen clonality of MMR-deficient gliomas relative to other MMR-deficient cancers may explain the lack of both immune recognition and response to PD-1 blockade in gliomas. Our data suggest a change in practice whereby tumor re-sequencing at relapse to identify progression and hypermutation could inform prognosis and guide therapeutic management