• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 163
  • 100
  • 42
  • 25
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 5
  • 4
  • 3
  • 3
  • 3
  • Tagged with
  • 433
  • 91
  • 89
  • 58
  • 55
  • 55
  • 38
  • 34
  • 34
  • 32
  • 30
  • 29
  • 26
  • 25
  • 24
  • 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.
371

Crosstalk entre la kinase LKB1 et l'arginine methyltransferase PRMT5 dans le cancer du sein / Crosstalk between the kinase LKB1 and the arginine methyltransferase PRMT5 in breast cancer

Lattouf, Hanine 24 November 2017 (has links)
La protéine arginine méthyltransférase 5 est la majeure arginine méthyltransférase de type II chez les mammifères, responsable de la génération de la majorité des arginines protéiques symétriquement diméthylées. Elle est impliquée dans divers processus oncogéniques tel que la progression tumorale et la croissance indépendante de l'ancrage. PRMT5 est surexprimée dans plusieurs cancers comme le cancer de l'ovaire, des poumons et du colon. Cependant, son expression dans le cancer du sein n'est pas assez étudiée. Dans ce projet de thèse, nous avons analysé l'expression de PRMT5 dans une cohorte de 440 tumeurs mammaires. Nos résultats montrent que son expression nucléaire est un facteur de bon pronostic, notamment dans les tumeurs ERa-positives. Nous avons aussi mis en évidence une corrélation entre PRMT5 et la sérine/thréonine kinase LKB1, suggérant un lien entre ces deux protéines. Plusieurs approches in vitro et in cellulo nous ont permis de démontrer que PRMT5 et LKB1 interagissent dans le cytoplasme des cellules mammaires épithéliales. Bien que PRMT5 soit incapable de méthyler LKB1, nous avons montré pour la première fois que PRMT5 est un substrat de cette kinase. Nous avons par la suite identifié les Thr132, 139 et 144 comme cibles de la phosphorylation, au niveau du tonneau TIM en N-terminal de PRMT5. La mutation des thréonines T139/144 en alanine diminue significativement l'activité de PRMT5, probablement suite à une perte de son interaction avec des protéines régulatrices comme MEP50, pICLn et RiOK1. De plus, la modulation de l'expression de LKB1 altère l'activité de PRMT5, témoignant d'un nouveau mécanisme de régulation médié par la phosphorylation identifiée / Protein arginine methyltrasferase 5 is the major type II arginine methyltransferase in humans. It symmetrically dimethylates arginine residues on target proteins in both the cytoplasm and the nucleus. PRMT5 was reported to be an oncoprotein implicated in anchorage independent growth and tumor progression. So far, it has been involved in various cancers such as ovarian cancer, lung cancer and colon cancer, but its expression pattern in breast cancer has not been deeply studied. In this thesis project, we analyzed PRMT5 expression in a cohort of 440 breast tumor samples and we found that its nuclear expression is a good prognosis factor, mainly in ERa-positive tumors. Interestingly, our clinical results analysis showed that PRMT5 expression is correlated with the serine/threonine kinase LKB1, suggesting a relationship between both proteins. Several in vitro and in cellulo approaches gave evidence that PRMT5 and LKB1 interact directly in the cytoplasm of mammary epithelial cells. Moreover, although PRMT5 is not able to methylate LKB1, we found that PRMT5 is a bona fade substrate for LKB1. We next identified Thr132, 139 and 144 residues as target sites for phosphorylation, located in the TIM barrel domain of PRMT5. Interestingly, the Thr139/144 mutation to alanine decreased drastically PRMT5 methyltransferase activity, probably due to the loss of PRMT5 interaction with regulatory proteins such as MEP50, pICLn and RiOK1. In addition, the modulation of LKB1 expression modifies PRMT5 enzymatic activity, highlighting a new regulatory mechanism mediated by the discovered posttranslational modification of this arginine methyltransferase
372

Étude de la régulation de la méthylation du récepteur aux œstrogènes de type alpha dans la carcinogenèse mammaire : rôle de la protéine kinase LKB1 / Regulation of estrogen receptor alpha methylation in breast carcinogenesis : involvment of the protein kinase LKB1

Bouchekioua-Bouzaghou, Katia 05 July 2012 (has links)
Parallèlement à leur action nucléaire, les œstrogènes exercent également des effets via une signalisation cytoplasmique par des mécanismes pas complètement élucidés. Nous avons mis en évidence la méthylation de ERα (mERα) sur arginine est l’évènement clé de la signalisation non génomique des œstrogènes dans les cellules tumorales mammaires. Cette méthylation entraîne la formation d’un complexe contenant ERα/SRc/PI3K/FAK qui active des cascades de phosphorylation régulant la prolifération cellulaire. La production d’un anticorps spécifique de la forme méthylée a permis de montrer que ERα est hyperméthylé dans 55% des tumeurs mammaires. Afin de comprendre les mécanismes de régulation de la méthylation, nous avons recherché de nouveaux partenaires impliqués dans ce processus. Au cours de ma thèse, j’ai montré que la protéine kinase LKB1 est impliquée dans la signalisation non génomique des œstrogènes. Dans les cellules MCF-7, les œstrogènes entraînent rapidement le recrutement de LKB1 au sein du complexe précédemment décrit. De plus, LKB1 est indispensable à la formation de ce macro complexe ainsi qu’à la phosphorylation de Bad en aval. En effet, par cette action, LKB1 participe au rôle protecteur des œstrogènes contre l’apoptose orchestré par les œstrogènes. De plus, une étude de l’expression de mERα et LKB1, sur une série de tumeurs mammaires, a montré une corrélation significative entre l’expression de ces deux protéines associée à l’envahissement ganglionnaire. Ces résultats révèlent une signification biologique de l’interaction LKB1/mERα, suggérant un rôle oncogénique putatif de LKB1 / Besides its nuclear action, estrogens mediate also cytoplasmic signaling, however the mechanisms are not fully understood. We recently showed that arginine methylation of estrogen receptor alpha (Erα) is required for the recruitment of PI3K and Src, activating downstream kinases. An antibody that specifically recognized Erα dimethylated was generated allowing the detection of Erα hypermethylation in 55% of breast tumors. To decipher the molecular mechanisms that regulate Erα methylation in non genomic pathways, we investigated new partners of the methylated form of Erα (mERα). We identified the tumor suppressor LKB1, a Ser/Thr kinase involved in cell metabolism and cell polarity as a new partner of mERα. To ascribe a biological role to this interaction, we analyzed the protein complexes containing mErα and LKB1. LKB1 is part of the complexe involved in Erα non genomic pathway. LKB1 is essential for Erα methylation and the formation of the macrocomplex Erα/p85/Src suggesting a functional role of LKB1 in Erα methylation and then activation of downstream signaling pathways. Using a phosphospecific antibody microarray, we observed that LKB1 was required for Bad phosphorylation, suggesting its involvement in apoptosis. Indeed, we found that LKB1 participes in the protective role of estrogen against apoptosis. Interestingly, an IHC study on human breast tumors points a correlation between the expression of LKB1 and mErα : their expression is correlated with lymph node metastasis. Altogether, these results reveal biological significance of mErα/LKB1 interaction, suggesting a putative oncogenic role to LKB1
373

Étude de la régulation de la méthylation du récepteur aux œstrogènes de type alpha dans le cancer du sein / Regulation of estrogen receptor alpha methylation in breast cancer

Poulard, Coralie 27 September 2013 (has links)
Le cancer du sein représente une cause de mortalité élevée chez la femme. Le cancer du sein est un cancer hormono-dépendant. De ce fait, il est extrêmement important de définir le rôle joué par les différents acteurs protéiques de la signalisation hormonale, notamment la signalisation œstrogénique. Parallèlement aux effets nucléaires de ERa où l'hormone lie le récepteur nucléaire et régule la transcription génique, il existe une voie dite non génomique. L'équipe a montré que les œstrogènes induisent la méthylation de ERa, qui est un prérequis au recrutement de la Pl3K et de la tyrosine kinase Src, conduisant à l'activation de molécules de signalisation telles que les MAPK et Akt, induisant prolifération et survie cellulaire. Durant ma thèse, j'ai pu démontrer que le complexe mERa/Src/Pl3K existe in vivo et constitue un nouveau biomarqueur indépendant de mauvais pronostique. La recherche de nouveaux partenaires du complexe mERa/Src/Pl3K nous a permis d'identifier le suppresseur de tumeur LKB1 et l'arginine déméthylase JMJD6. De façon surprenante, l'étude de l'expression de LKB1 par immunohistochimie dans une cohorte de tumeurs mammaires a montré une dualité fonctionnelle selon sa localisation subcellulaire. De plus, nous avons démontré que JMJD6 s'associe à ERa méthylé lorsque le récepteur est complexé à Src et Pl3K, et permet ainsi la déméthylation de ERa et la dissociation du complexe mERa/Src/Pl3K. Ce travail a ainsi pu mettre en évidence que les différents acteurs de cette signalisation peuvent constituer des éléments clés au diagnostique mais également lors de la décision thérapeutique, puisque qu'il existe des drogues peuvant cibler cette voie de signalisation / Estrogen receptor a {ERa}, belonging to the superfamily of hormone nuclear receptors, regulates many physiological processes, notably the growth and survival of breast tumor cells, acting as a ligand-dependent transcription factor. Besides to the well described transcriptional effects, estrogen also mediate extranuclear events called non genomic signaling via its receptor. /n fact, team shows that ERa is methylated and that this event is a prerequisite for the recrutement of Src and P/3K and the activation of Akt which orchestrate cell proliferation and survival. During my PhD, / demonstrated that the non genomic signaling complex mERa/Src/P/3K exists in vivo and is operative. /n addition, the complex is found to be an independent prognostic factor for disease free survival. This is an emergent concept that estrogen non genomic pathway is operative in vivo and can constitute a new therapeutic targets. The search for new partners of the complex has allowed us to identify the tumor suppressor LKB1 and arginine demethylase JMJD6. Expression of LKB1 in immunohistochemistry revealed dual properties based on its subcellular localization. When LKB1 is complexed with mERa/Src/P/3K it may acquire oncogenic properties. /n addition, JMJD6 interacts with methylated ERa when the receptor is associated with Src and P/3K, and allows the demethylation of ERa and the dissociation of the complex mERa/Src/P/3K. This work showed that estrogenic non genomic players can constitute new therapeutic targets in Breast tumors
374

Mechanism-Based Personalized Medicine for Cystic Fibrosis by Suppressing Pseudo Exon Inclusion / 偽エクソン生成を標的とした嚢胞性線維症に対する個別化医療

Shibata, Saiko 23 March 2021 (has links)
京都大学 / 新制・課程博士 / 博士(医学) / 甲第23065号 / 医博第4692号 / 新制||医||1049(附属図書館) / 京都大学大学院医学研究科医学専攻 / (主査)教授 村川 泰裕, 教授 平井 豊博, 教授 小川 誠司 / 学位規則第4条第1項該当 / Doctor of Medical Science / Kyoto University / DFAM
375

PRMT1, un nouveau corégulateur de la signalisation de la progestérone dans le cancer du sein / PRMT1, un nouveau corégulateur de la signalisation de la progestérone dans le cancer du sein

Malbéteau, Lucie 11 October 2019 (has links)
La progression du cancer du sein repose principalement sur la signalisation des œstrogènes et de la progestérone, et les traitements modulant l’action des œstrogènes ont amélioré la survie des patientes atteintes d’un cancer à récepteurs œstrogéniques (ERα). Des études récentes convergent sur le concept selon lequel, dans les cancers du sein ER+, PR (Progesterone Receptor) peut inhiber les fonctions favorisant la croissance induite par l'œstrogène en reprogrammant directement la liaison d'ERα sur de nouveaux gènes cibles. Les données cliniques montrent que cette signature génique est associée à un bon pronostic dans une cohorte de 1.959 patientes atteintes de cancer du sein et qu’un agoniste de la progestérone améliore l'activité antiproliférative des thérapies anti-oestrogéniques1. Ainsi, ces données démontrent qu’ER n’est pas le seul acteur de la tumorigénèse mammaire et qu'il existe une interférence fonctionnelle entre ces deux voies hormonales, soulignant le besoin d’une meilleure compréhension de la signalisation de PR. D’un point de vue mécanistique, l’activité de PR est étroitement liée à l’interaction avec les nucléosomes. En effet, PR fonctionne comme un facteur « pionnier » et se lie à la chromatine au sein de complexes protéiques, régulant son activité transcriptionnelle. Sans progestérone, PR forme un complexe répressif associé à des enzymes modificatrices de la chromatine comme LSD1, HDAC1/2 et la protéine de l'hétérochromatine HP1γ2. En réponse au traitement hormonal, ce complexe est déplacé, ce qui permet de recruter des coactivateurs et des cofacteurs associés, qui modifient la structure de la chromatine locale et entraînent l'activation ou la répression des gènes cibles de PR. Nous avons identifié un nouveau régulateur de la signalisation de la progestérone, l'arginine méthyltransférase PRMT1, enzyme souvent surexprimée dans les cancers mammaires3,4. Par diverses approches in vitro et in vivo, nous avons montré une interaction directe entre PR et PRMT1, dans le noyau des cellules tumorales mammaires, et à la fois en absence d’hormone et après 1h de stimulation à la progestérone. De plus, PRMT1 apparaît comme un nouveau membre du complexe répressif sur la chromatine, associé à PR et à ses partenaires, dans un sous-ensemble de gènes inductibles par la progestérone. Nos résultats indiquent également que l’expression de PRMT1 affecte l’activité transcriptionnelle de PR et que son inhibition perturbe l’activation rapide de la voie de la protéine kinase après une stimulation progestative. Nous montrons pour la première fois que PR est méthylé sur un résidu arginine, conservé parmi les récepteurs nucléaires (R637), localisé dans son domaine de liaison à l'ADN. La production d’un anticorps dirigé contre la forme méthylée de PR nous a permis de préciser qu’elle se localisait dans le noyau des cellules et n’était retrouvée qu’après traitement progestatif. En outre, la mutation de R637 de PR entraine une diminution de l’expression d'un sous-ensemble de cibles de PR, ce qui entraine un retard de croissance cellulaire. En conclusion, ces résultats confirment l'implication de PRMT1 et de son activité méthyltransférase dans la signalisation de PR et plus particulièrement dans son activité transcriptionnelle. Nous démontrons donc que la méthylation sur résidus d'arginine est un nouveau mécanisme de contrôle lors de la réponse à la progestérone dans les cellules tumorales mammaires / Breast cancer progression is mainly driven by estrogen and progesterone signalling and therapies modulating oestrogen‘s action have improved the survival of ER+ cancer patients. As progesterone receptor (PR) is an ER target gene, its expression in breast cancer was considered as a predictive marker of ER functionality. However, recent studies are converging on the concept that PR can directly affect ER functions in breast cancer cells1. Activated PR can redirect ER to novel chromatin binding sites associated with cell differentiation and apoptosis, leading to a potential improvement of the tumour response to anti-oestrogen therapies. In considering the differential effects of progesterone in breast cancer, it is important to define the variable might influence progesterone pathway and the downstream mediators involved in this signalling. Recently, Beato and al reported that, in breast cancer cells, the unliganded form of PR (non-activated with progesterone) bind to genomic sites and target a repressive complex containing enzyme modifying chromatin as the demethylase LSD1 or the Heterochromatin Protein 1 (HP1γ)2. Under hormonal treatment, this complex is displaced, which makes it possible to recruit coactivators and associated cofactors, which modify the structure of the local chromatin and cause the activation or repression of the target genes of PR. In addition, cellular response to progesterone is also regulated by receptor post-translational modifications that may affect its stability, its subcellular localization and its interactions with regulators. In our study, we demonstrated for the first time that PR is methylated on arginine residues, by the arginine methyltransferase PRMT1. We identified as target the arginine 637 (R637), a conserved arginine among nuclear receptor superfamily, located in the DNA-binding domain of the receptor. By in vitro and in vivo approaches, we are studying the impact of PRMT1 on PR signalling pathways. In T47D breast cancer cells, we demonstrated that PR interacts with PRMT1, mainly in the nucleus. Of interest, PRMT1 interacts with PR in the nucleus in absence of hormone stimulation and it appears as a new member of the repressive complex on a subset of progesterone inducible genes. Our results also indicate that PRMT1 expression affects PR transcriptional activity and PRMT1 knockdown disrupts the rapid activation of protein kinase pathway after progestin stimulation. The production of an antibody directed against the methylated form of PR allowed us to specify that methylated-PR is localized in the nucleus of cells and was found only after progesterone treatment. Furthermore, PRMT1 depletion and mutation of R637 resulted in an inhibition of a subset of PR-regulated genes which led to retarded cell growth.Our data reveal the impact of PRMT1 expression on PR pathways and provide evidence for the asymmetric arginine dimethylation of PR. We therefore demonstrate that methylation on arginine residues could be a novel control mechanism in the response to progesterone in mammary tumor cells
376

Autosomal Dominant Neurohypophyseal Diabetes Insipidus in Two Families: Molecular Analysis of the Vasopressin-Neurophysin II Gene and Functional Studies of Three Missense Mutations

Hedrich, Christian Michael, Zachurzok-Buczynska, Agnieszka, Gawlik, Aneta, Russ, Susanne, Hahn, Gabriele, Köhler, Katrin, Malecka-Tendera, Ewa, Hübner, Angela January 2009 (has links)
Background: Autosomal dominant familial neurohypophyseal diabetes insipidus (adFNDI) is a rare disease with symptoms of polydipsia, polyuria and dehydration caused by arginine vasopressin deficiency. Disease onset is within infancy or adolescence. A variety of disease-causing mutations of the arginine vasopressin neurophysin II gene (AVP) on chromosome 20p13 have been described. Methods: Two Polish families with adFNDI were screened for mutations. Processing of wild-type (WT) and mutant AVP was monitored using immunocytochemical methods in stably transfected Neuro2A cells. AVP secretion into the cell culture supernatant was investigated with an enzyme immunoassay. Results: In the first family a heterozygous p.G96D mutation was identified. Some patients additionally carried a novel heterozygous mutation p.A159T. The second family presented with a heterozygous mutation p.C98G. Confocal laser microscopy unveiled accumulation of p.G96D and p.C98G prohormones in the cellular bodies, whereas WT and p.A159T prohormones and/or processed products were located in the tips of cellular processes. Reduced levels of AVP in supernatant culture medium of p.G96D and p.C98G transfected cells in comparison to p.A159T and WT cells were found. Conclusions: We conclude that the p.G96D and p.C98G mutations cause adFNDI in the two reported families. The sequence variant p.A159T does not seem to have disease-causing effects. / Dieser Beitrag ist mit Zustimmung des Rechteinhabers aufgrund einer (DFG-geförderten) Allianz- bzw. Nationallizenz frei zugänglich.
377

Roles of Protein Arginine Methyltransferase 7 and Jumonji Domain-Containing Protein 6 in Adipocyte Differentiation: A Dissertation

Hu, Yu-Jie 28 October 2015 (has links)
Regulation of gene expression comprises a wide range of mechanisms that control the abundance of gene products in response to environmental and developmental changes. These biological processes can be modulated by posttranslational modifications including arginine methylation. Among the enzymes that catalyze the methylation, protein arginine methyltransferase 7 (PRMT7) is known to modify histones to repress gene expression. Jumonji domain-containing protein 6 (JMJD6) is a putative arginine demethylase that potentially antagonize PRMT7. However, the biological significance of these enzymes is not well understood. This thesis summarizes the investigation of both PRMT7 and JMJD6 in cell culture models for adipocyte differentiation. The results suggest that PRMT7 is not required for the differentiation, whereas JMJD6 is necessary for the differentiation by promoting the expression of the lineage determining transcription factors peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancerbinding proteins (C/EBPs). The underlying mechanisms by which JMJD6 regulate differentiation involve transcriptional and post-transcriptional control of gene expression. Unexpectedly, the adipogenic function of JMJD6 is independent of its enzymatic activity. Collectively, the present research reveals a novel role of JMJD6 in gene regulation during the differentiation of adipocytes.
378

Investigation of Protein Dynamics and Communication in Adomet-Dependent Methyltransferases: Non-Ribosomal Peptide Synthetase and Protein Arginine Methyltransferase

May, Kyle M. 01 August 2019 (has links)
For many enzymes to function correctly they must have the freedom to display a level of dynamics or communication during their catalytic cycle. The effects that protein dynamics and communication can have are wide ranging, from changes in substrate specificity or product profiles, to speed of reaction or switching activity on or off. This project investigates the protein dynamics and communication in two separate systems, a non-ribosomal peptide synthetase (NRPS), and a protein arginine methyltransferase (PRMT). PRMT1, the enzyme responsible for 80% of arginine methylation in humans, has been implicated in a variety of disease states when functioning incorrectly. For this reason, much focus has been placed on better understanding how PRMT1 determines which products it creates and at what times. This project aims to shed light on how dynamics and communication within PRMT1 dictate its activity. We have to this point developed a protocol for creating and purifying a linked PRMT1 construct which will enable us to conduct the necessary experiments capable of answering our larger questions about the PRMT1 catalytic mechanism. Our collaborators in the Zhan lab discovered the presence of a methyltransferase (Mt) in the two NRPS systems they study, which produce two different and medically relevant compounds, bassianolide and beauvericin. The Hevel lab is well suited to study methyltransferases and so were asked to help evaluate the role of these Mt domains and how they affect the production of the relevant natural products. Achieving a more complete understanding of these systems will move us closer toward the “holy grail” of being able to manipulate and harness NRPS systems for the engineering of novel medically relevant compounds. This project has found that the Mt domain substrate specificity is affected by the surrounding protein domains, or even small portions of them.
379

Interakce hyaluronan-aminokyseliny / Hyaluronan-amino acids interactions

Jugl, Adam Unknown Date (has links)
The presented dissertation focuses on non-covalent interactions of hyaluronan of different molecular weights (9–1540 kDa) with basic (oligo)-amino acids (especially arginine) and the antimicrobial peptide cecropin B. High-resolution ultrasonic spectroscopy (HR-US), isothermal titration calorimetry (ITC) and potentiometric titration techniques were chosen to investigate the interactions. The thesis focuses on the characterization of interactions, especially with respect to the used molecular weight of interacting polymers and the ionic strength of the environment. Whether interactions occur or not was determined primarily by the length of the arginine oligomer chain. For monomeric amino acids, the interactions were investigated mainly by potentiometric titrations. Interactions were observable from arginine oligomers with eight monomer units. The molecular weight of hyaluronan mainly affected the intensity of the interactions. The transition between the individual conformations of hyaluronan (rod and random coil) was especially significant. Investigation of interactions was performed in water, in solutions with different concentrations of sodium chloride and in PBS. The sufficiently high ionic strength of the solution was able to suppress the interactions in water between the oligomers of arginine and hyaluronan. The basic antimicrobial peptide cecropin B has been shown to interact with hyaluronan in water but not in PBS. Based on these results, it was possible to conclude that the hyaluronan-cecropin B system would be particularly suitable for topical applications.
380

Ribonucleoprotein complexes and protein arginine methylation : a role in diseases of the central nervous sytem

Chénard, Carol Anne. January 2008 (has links)
No description available.

Page generated in 0.0477 seconds