• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 8
  • 7
  • 6
  • 4
  • 2
  • 1
  • 1
  • Tagged with
  • 38
  • 38
  • 16
  • 14
  • 14
  • 12
  • 7
  • 7
  • 7
  • 5
  • 5
  • 5
  • 5
  • 4
  • 4
  • 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.
21

Willin as a novel 4.1 ezrin radixin moesin (FERM) domain protein in the mammalian Hippo signalling pathway

Angus, Liselotte January 2011 (has links)
The Salvador/Warts/Hippo (Hippo) pathway defines a novel signalling cascade regulating cell contact inhibition, organ size control, cell growth, proliferation, apoptosis and cancer development in mammals. The Hippo pathway was initially utilised in D. melanogaster, where the Expanded protein acts in the Hippo signalling cascade to control organ size. Willin is the proposed human orthologue of Expanded and the aim of this thesis is to investigate whether willin can activate the mammalian Hippo signalling pathway. Ectopic willin expression causes an increase in phosphorylation of the core Hippo signalling pathway components MST1/2, LATS1 and YAP, an effect which can be antagonised by ezrin. In MCF10A cells, willin over-expression antagonises a YAP-induced epithelial-to-mesenchymal transition via the N- terminal FERM (Four-point-one Ezrin Radixin Moesin) domain of willin. Preliminary results show that willin is expressed within the sciatic nerve of rat and mice, and within the neuromast cells in the zebrafish; suggesting that willin and the Hippo pathway may play a vital role in the developmental regulation within the peripheral nervous system. To conclude, willin influences Hippo signalling activity by activating the core Hippo pathway kinase cassette in mammalian cells.
22

Investigation of Altered Cell-Cell Interactions and Signaling Mechanisms in <i>Drosophila</i> Tumor Models

Waghmare, Indrayani 08 September 2016 (has links)
No description available.
23

Etude des mécanismes moléculaires régulant la voie Hippo via les intégrines ß1 / Study of the molecular mechanisms regulating the Hippo pathway via the integrins b1

Sabra, Hiba 29 June 2017 (has links)
L'adhérence cellulaire à la matrice extracellulaire joue un rôle clé dans leur prolifération,leur différenciation ou l'apoptose. Par conséquent ce processus est critique pour undéveloppement normal et pour l'homéostasie tissulaire. La dérégulation de ce mécanismecontribue souvent à des situations pathologiques. Ainsi, la dérégulation de nombreux gènesimpliqués dans les adhérences cellule-cellule ou cellule-matrice extracellulaire sont liés à despathologies conduisant à un défaut de développement, la progression tumorale, oul'inflammation.Les intégrines sont des récepteurs transmembranaires hétéro dimériques jouant un rôlemajeur dans les interactions cellule-matrice extracellulaire. Ce rôle n'est pas limité à unesimple interaction mécanique puisqu'elles permettent également la transduction dessignaux de la matrice extracellulaire à la cellule afin de permettre à cette dernière des'adapter à son micro environnement. Dans le but d’étudier le rôle des intégrines à chaîneβ1 dans le développement osseux, le laboratoire a mis en place un modèle murind'inactivation conditionnelle du gène Itgb1 basée sur l'expression de la recombinase Cre austade pré-ostéoblastique. Les souris mutées présentent un défaut de développementosseux, dû à une faible prolifération des ostéoblastes.Contrairement à ce qui était généralement admis, cette faible prolifération desostéoblastes est indépendante de la voie classique mettant en jeu la voie classique des MAPkinases. En revanche, elle est contrôlée par la voie Hippo: cette signalisation a étérécemment identifiée chez la Drosophile et les Mammifères comme un mécanismeinhibiteur majeur de la prolifération cellulaire. Le cofacteur de transcription YAP, effecteurfinal de cette voie, est une navette nucléo-cytoplasmique. Son expression est amplifiée dansdivers cancers dont l'ostéosarcome où cette surexpression associée à celle de l’Itgb1 est unfacteur de mauvais pronostique.Mes travaux consistent à comprendre comment les intégrines à chaîne β1 contrôlent lavoie Hippo, et donc la prolifération. Nous avons confirmé que la délétion des intégrines β16active la phosphorylation de YAP et sa séquestration dans le cytoplasme. En utilisant destechniques de Biologie Cellulaire et de Biochimie, nous avons montré que suite à la délétionde l’Itgb1, les cellules présentent un défaut de trafic vésiculaire réduisant la translocationmembranaire de Rac1. La séquestration cytoplasmique de Rac1 diminue l’activation de soneffecteur majeur la kinase PAK responsable de la dissociation d'un complexe membranaired'inactivation composé de la protéine adaptatrice NF2, la kinase LATS et de son effecteurprincipal YAP. Les intégrines en provocant la perte de ce complexe induisent ladéphosphorylation de YAP, sa translocation nucléaire et donc stimulent la proliférationcellulaire. / Cell adhesion to the extracellular matrix plays a key role in their proliferation,differentiation or apoptosis. Therefore, this process is critical for normal development andtissue homeostasis. The deregulation of this mechanism often contributes to pathologicalsituations. Thus, the deregulation of many genes involved in cell-cell or cell-extracellularmatrix adhesions are linked to pathologies leading to developmental defects, tumorprogression, or inflammation.Integrins are heterodimeric transmembrane receptors that play a major role in cellextracellularmatrix interactions. This role is not limited to a simple mechanical interactionsince integrins also allow the transduction of the signals from the extracellular matrix to thecell in order to permit the latter to adapt to its microenvironment. In order to study the roleof β1 integrins in bone development, the laboratory has implemented a mouse model withconditional inactivation of the Itgb1 gene based on the expression of recombinase Cre at thepre-osteoblastic stage. The mutated mice show a defect in bone development due to a lowproliferation rate of osteoblasts.Contrary to what was generally accepted, this reduced proliferation is independent of theclassical pathway involving the classical pathway of MAP kinases. On the other hand, it iscontrolled by Hippo: this signaling pathway has recently been identified in Drosophila andMammals as a major inhibitory mechanism of cell proliferation. The transcription cofactorYAP, the end effector of this pathway, is a nucleo-cytoplasmic shuttle. Its expression isamplified in various cancers including osteosarcoma where this overexpression associatedwith that of Itgb1 is a factor of poor prognosis.My work involves understanding how β1 integrins control the Hippo pathway, and thusproliferation. We confirmed that deletion of β1 integrins activates the phosphorylation ofYAP and its sequestration in the cytoplasm. Using Cell Biology and Biochemistry techniques,we showed that following the deletion of Itgb1, the cells exhibit a defect in vesicular trafficthat reduces the membrane translocation of Rac1. The cytoplasmic sequestration of Rac18decreases the activation of its major effector, the PAK kinase. PAK is responsible for thedissociation of an inactivating membrane complex composed of the adaptor protein NF2,the LATS kinase, and its main effector YAP. The integrins by provoking the loss of thiscomplex induce the dephosphorylation of YAP, its nuclear translocation, and thus stimulatecell proliferation.
24

Characterization of the pathophysiological mechanisms associated with NEK8/NPHP9 mutations identified in patients with severe renal ciliopathies / Caractérisation des mécanismes physiopathologiques associés aux mutations NEK8 / NPHP9 identifiées chez des patients atteints de ciliopathies rénales sévères

Grampa, Valentina 21 September 2015 (has links)
Les ciliopathies sont un groupe de maladies génétiques multi-systémiques liées à un dysfonctionnement du cil primaire, une structure sensorielle présente à la surface des cellules qui régule des voies de signalisation clés au cours du développement et de l'homéostasie tissulaire. Afin d'identifier de nouveaux gènes responsables de ciliopathies développementales sévères, ~ 500 patients / fétus ont été analysés par une approche de séquençage à haut débit de l'exome ciblant > 1 200 gènes ciliaires ("ciliome"). Nous avons identifié huit nouvelles mutations dans le gène NEK8/NPHP9 chez cinq familles dont les syndromes se chevauchent. NEK8/NPHP9 code une protéine kinase de la famille des NIMA qui se localise au niveau du compartiment Inversine du cil primaire et agit comme un régulateur de la signalisation Hippo, une voie essentielle contrôlant la taille des organes. Nous montrons pour la première fois que les mutations du gène NEK8 sont associées à une agénésie rénale et une hypodysplasie. De plus, notre travail met en évidence une corrélation génotype/phénotype: les mutations "perte de fonction" de NEK8 conduisant à reins élargies et kystiques, des kystes pancréatiques et hépatique, alors que les mutations faux-sens de NEK8 causent une hypodysplasie/agénésie rénale associée à une cardiopathie et une paucité des canaux biliaires. La première partie de mon projet de thèse porte sur l'étude de l'impact des mutations faux-sens de NEK8 sur divers processus cellulaires et des voies de signalisation dépendantes de NEK8. Nous avons démontré un effet "gain de fonction" des mutations faux-sens de NEK8 puisqu'elles affectent la ciliogenèse et la composition du compartiment Inversine (localisation ciliaire de ANKS6). De plus, ces mutations altèrent la localisation nucléaire de YAP, le principal acteur de la voie Hippo, ainsi que l'expression des gènes cibles de YAP dans les fibroblastes de patients et dans la lignée cellulaire rénale (mIMCD3) invalidée pour NEK8. De même, nous avons montré une accumulation anormale de YAP nucléaire dans les reins polykystiques de la souris Jck, porteuse d'une mutation faux-sens de Nek8. Un déséquilibre de la voie Hippo serait donc à l'origine des défauts de morphogenèses épithéliales. En effet, les cellules mIMCD3 invalidées pour NEK8 forment en culture 3D des structures anormales et/ou des sphères élargies qui s'accompagnent d'une persistance du marquage nucléaire de YAP et Ki-67 et forment de grandes sphères par rapport aux cellules contrôles. Des défauts plus sévères ont été observés pour les cellules ré-exprimant les différents mutants de NEK8, confirmant la pathogénicité de ces mutations et leur effet "gain de fonction". Enfin, le traitement par la Vertéporfine, un inhibiteur spécifique de l'activité transcriptionnelle de YAP, améliore non seulement le phénotype des fibroblastes de patients et des cellules rénales invalidées pour NEK8 en culture 3D, mais également in vivo les anomalies observées chez les embryons de poisson zèbre dues à la surexpression de la forme NEK8 humaine, confirmant ainsi l'implication d'une dérégulation de YAP dans les mécanismes physiopathologiques. Par ailleurs, nous avons observé que les mutants de NEK8 s'accumulent de manière anormale au niveau de l'appareil de Golgi dans les fibroblastes de patients, et que cet appareil de Golgi apparait dispersé. Nos résultats montrent que le recrutement de NEK8 au Golgi est sensible à la Brefeldine A et dépendrait donc de ARF1, une petite GTPase impliquée dans le trafic de protéines entre les compartiments du Golgi et du réticulum endoplasmique. Nous avons démontré que NEK8 interagit et co-localise préférentiellement avec la forme d'ARF1 liée au GDP, suggérant pour NEK8 une possible fonction de facteur d'échange d'ARF1 à des sites spécifiques (appareil de Golgi, membranes, cil) afin de promouvoir le trafic vésiculaire de protéines telles que les protéine ciliaires. (...) / Ciliopathies are a group of genetic multi-systemic disorders related to dysfunction of the primary cilium, a sensory organelle present at the cell surface that regulates key signaling pathways during development and tissue homeostasis. In order to identify novel genes whose mutations would cause severe developmental ciliopathies, ~500 patients/fetuses were analyzed by a targeted high throughput sequencing approach allowing exome sequencing of > 1200 ciliary genes. We have identified eight novel mutations in NEK8/NPHP9 in five independent families with severe overlapping syndromic disorders. NEK8/NPHP9 encodes a NIMA-related kinase that localizes at the inversin compartment of the primary cilium and acts as a regulator of Hippo signaling, a pathway that is essential for control of organ size during development. We show for the first time that NEK8 mutations are associated with renal agenesis and hypodysplasia, and our work highlights a genotype/phenotype correlation with NEK8 loss-of-function mutations leading to enlarged cystic kidney, pancreas and liver, whereas NEK8 gain-of-function (missense) mutations cause renal hypodysplasia, cardiopathy and paucity of bile ducts. The first part of my thesis project focuses on the study of the impact of these NEK8 missense mutations on various cellular processes and NEK8-dependent signaling pathways. We demonstrate that NEK8 missense mutations impair the Inversin (INVS) compartment composition and ciliogenesis, and also alter the nuclear localization of the main Hippo signalling effector, YAP, as well as expression of its target genes in patient fibroblasts and renal cell lines. We also demonstrated that this Hippo pathway imbalance causes epithelial morphogenesis defects in 3D matrigel culture. Indeed, mIMCD3 cells depleted for NEK8 showed persistent YAP and Ki-67 staining and formed bigger spheres compared to control cells. Abnormal sphere volume was also observed in cells re-expressing NEK8-GFP mutations, suggesting their pathogenicity. We confirm these data in vivo in Jck mice, a model of polycystic kidney disease bearing a Nek8 missense mutation. Finally, treatment with Verteporfin, a specific inhibitor of YAP transcriptional activity, improves the mutant phenotype of both cellular models and zebrafish embryos overexpressing human NEK8, further supporting the involvement of YAP dysregulation in the pathogenic cellular mechanisms. Surprisingly, in patient fibroblasts, we showed that mutated NEK8 accumulates at the Golgi that appeared dispersed. NEK8 recruitment at the Golgi apparatus is dependent on ARF1 (Brefeldin A sensitive), a small GTPase involved in protein trafficking between Golgi compartments and ER. We notably demonstrated that NEK8 mostly interacts and localizes with the dominant negative form of ARF1 (T31N), suggesting that NEK8 could act as an activator (GEF) of ARF1 to promote vesicular trafficking of ciliary proteins. The second part of my project focuses on a new candidate gene for which a missense homozygous mutation has been identified in 3 individuals presenting a late onset NPH with hepatic fibrosis. This gene encodes ANKS3, an evolutionarily conserved protein whose function is still poorly characterized. Interestingly, ANKS3 has been reported to be a partner of NEK8, even though we showed it does not localize at the INVS compartment with NEK8 but is rather present at the base of cilia in fibroblasts. We showed that the missense mutation does not affect ANKS3 localization but leads to longer cilia and abnormal accumulation of NEK8 at the cilium base in patient fibroblasts and kidney tubules. Altogether, my work focused on NEK8 and its partners, ANKS6 and ANKS3, each of whose related gene is mutated in patients presenting a broad clinical spectrum of phenotypes. (...)
25

Drosophila Eye Model to Study Dorso-Ventral (DV) Patterning and Neurodegenerative Disorders

Gogia, Neha January 2019 (has links)
No description available.
26

Maternal Hepatic Adaptations to Pregnancy

Shashank Manohar Nambiar (11177052) 06 August 2021 (has links)
<p>During gestation, the maternal liver undergoes various adaptive changes to cope with the increasing physiological and metabolic demands from both maternal and fetal compartments. Among these changes are robust growth and changes in transcriptome profile. However, how these events happen, and other aspects of this physiological phenomenon remains unexplored. Therefore, we aimed at further understanding how maternal liver responds to pregnancy. We used BrdU labeling combined with a virus-based tracing approach to quantify the percentage of maternal hepatocytes undergoing DNA synthesis and division over the course of gestation in mice. </p> <p>We found that ~50% maternal hepatocytes entered S-phase but, unexpectedly, did not undergo cytokinesis. This strongly suggests that maternal hepatocytes in fact undergo endoreplication instead of hyperplasia, as believed previously. Pericentral Axin2<sup>+</sup> hepatocytes were reported to behave as liver stem cells responsible for liver homeostasis and turnover. We generated an <i>in vivo</i> fate-tracing mouse model to monitor the behavior of these cells in the maternal liver. Our results showed that they did not proliferate during pregnancy, homeostasis, and following partial hepatectomy. Curiously, we uncovered that, hepatocytes exhibit developmental phenotypes at mRNA level pre-pregnancy and at both mRNA and protein level during pregnancy. In the non-pregnant state, hepatocytes reserved mRNA expression of liver progenitor marker genes <i>Cd133</i> and <i>Afp</i>, which are localized in the nuclei, without protein translation. During gestation, maternal hepatocytes displayed cytoplasmic translocation of <i>Cd133</i> and <i>Afp</i> transcripts, concomitant with corresponding protein expression. </p> <p>Overall, all maternal hepatocytes became CD133<sup>+</sup>, and a subset of them express AFP. Additionally, in non-pregnant livers, mRNA of <i>Epcam</i>, another liver progenitor marker, was expressed within majority of hepatocytes, whereas its protein was solely translated in the pericentral region. In contrast, by end-gestation, EPCAM protein expression switched to the periportal region. These observations indicate that maternal hepatocytes exhibit heterogeneous developmental phenotypes, partially resembling fetal hepatocytes. It is intriguing why mature hepatocytes dedifferentiate into a progenitor state in response to pregnancy. AFP is considered to be produced primarily from fetal liver and thus is used to evaluate fetal development health. </p> A potential clinical relevance of our data is that we identified maternal liver as a new source of AFP. The hippo signaling pathway has been shown to potently control liver growth and hepatocyte heterogenicity. Surprisingly, we found that pregnancy neither altered the expression nor activities of the components of this pathway and its effector YAP1/TAZ. This finding indicates that pregnancy-induced maternal liver growth is not driven by hippo-YAP1 pathway. However, we demonstrate that the presence of YAP1 is essential for CD133 protein expression in maternal hepatocytes. Collectively, we revealed that, as pregnancy advances, maternal hepatocytes likely undergo endoreplication and display developmental phenotypes. Mechanistically, YAP1 dictates the expression of CD133, contributing to the pregnancy-dependent phenotypic changes of maternal hepatocytes.
27

Regulation of Dronc Transcription by the Hippo and Ecdysone Pathways in Drosophila Melanogaster

Gangwani, Karishma 11 August 2022 (has links)
No description available.
28

Using BioID to study RAS signaling to the Hippo pathway

Nikolova, Maya 08 1900 (has links)
RAS est une GTPase qui transduit les signaux extracellulaires envers des voies de signalisation intracellulaires, en liant ses effecteurs. RAS peut activer la voie Hippo qui inhibe la croissance cellulaire et qui est souvent dérégulée dans le cancer. Les protéines RASSF suppresseurs de tumeurs relient RAS à la voie Hippo. L’expression exogène de KRASG12V avec RASSF1 ou RASSF5 conduit à l'activation de la voie Hippo, bien que KRAS et RASSF1 ne s’associent pas directement. Ce projet de maîtrise vise à identifier les protéines impliquées dans l'activation de la voie Hippo par RAS. Nous avons effectué plusieurs expériences BioID, une technique qui permet d’identifier les interacteurs proximaux d’une protéine d’intérêt, dans des lignées cellulaires U2OS stables et inductibles exprimant les protéines KRASG12V, RASSF1 ou RASSF5 seules ou coexprimées, permettant de comparer les conditions où la voie Hippo inactive ou active. Nous avons élucidé l'interactome d'un mutant de KRAS avec affinité accrue envers RASSF5 et affinité réduite envers RAF, permettant d’étudier les voies activées en aval de RASSF5, avec une activation réduite de la voie MAPK. Nos données montrent que RASSF1 et RASSF5 relâchent les kinases Hippo MST1 et MST2 lorsque la voie Hippo est active, conformément aux données in vitro démontrant un rôle inhibiteur de l'interaction RASSF/MST. De plus, nous avons démontré que KRAS est un interacteur proximal des protéines VAMP3 et SNAP23. Comprendre comment l'oncoprotéine RAS active des effecteurs et des voies de signalisation moins étudiés, en particulier ceux qui ont des fonctions suppressives de tumeurs a des implications importantes pour le développement de nouvelles thérapies ciblées pour les cancers induits par RAS. / RAS is a small GTPase that transduces signals from membrane-bound receptors to intracellular pathways, by signaling to downstream effector proteins. RAS can activate the Hippo pathway, a growth-suppressive pathway that is often dysregulated in cancer. The tumor suppressor RASSF proteins link RAS to Hippo signaling. Co-expression of KRASG12V with either RASSF1 or RASSF5 leads to Hippo pathway activation, despite KRAS and RASSF1 not being direct binding partners. This M.Sc. project aims to identify proteins that are involved in RAS-mediated activation of the Hippo pathway. We performed BioID, a proteomic technique which is used to identify the proximal interactors of a protein of interest, in stable and inducible U2OS osteosarcoma lines expressing KRASG12V, RASSF1, or RASSF5 proteins alone, as well as in lines co-expressing both KRASG12V and the RASSF proteins, allowing for a comparison between inactive and active Hippo pathway interactomes. Furthermore, we mapped the interactome of a double mutant of KRAS that displays increased affinity for RASSF5 and decreased affinity for the effector RAF, allowing us to study KRAS signaling downstream of RASSF5, with decreased activation of the MAPK pathway. Our BioID data shows that RASSF1 and RASSF5 disengage the Hippo kinases MST1 and MST2 when the Hippo pathway is active, in line with the inhibitory role of the RASSF/MST interaction observed in vitro. Furthermore, we show that KRAS is a proximal interactor of the SNARE proteins VAMP3 and SNAP23. Understanding how the oncoprotein RAS signals to less studied effectors and pathways, particularly those with tumor suppressive functions has significant implications for understanding oncogenesis, and for development of new targeted therapies for RAS-driven cancers.
29

Expressão dos genes da via de sinalização celular hippo e aurora quinases na leucemia mielóide crônica / Expression of hippo signaling pathway and aurora kinase gene in chronic myeloid leukemia

Marsola, Ana Paula Zambuzi Cardoso 07 May 2018 (has links)
A Leucemia Mielóide Crônica (LMC) é uma neoplasia mieloproliferativa resultante da expansão clonal de células mielóides positivas para o cromossomo Philadelphia. A patogênese da LMC está associada à expressão do oncogene BCR-ABL1, que codifica a proteína Bcr-Abl com constitutiva atividade da tirosina quinase, promovendo a mieloproliferação exacerbada e a resistência à apoptose das células leucêmicas. Os pacientes com LMC são tratados principalmente com inibidores de tirosina quinase (TKI), mas a resistência aos inibidores e a refratariedade tem sido relatada em alguns pacientes na fase crônica e na maioria dos pacientes em fases avançadas da doença. Assim sendo, continua a ser de suma importância a elucidação da patogênese da LMC e a busca de novos alvos terapêuticos, como os membros da via de sinalização Hippo e reguladores do ciclo celular, da família Aurora quinase. O presente estudo quantificou o nível de expressão de genes que codificam componentes da via de sinalização Hippo (MST1, MOB1B, MOBKL1B, LATS1, LATS2, YAP e TAZ) e Aurora quinases A e B em: 1) pacientes com LMC em diferentes fases da doença, resistentes ou sensíveis à terapia com mesilato de imatinibe (MI), em indivíduos saudáveis e 2) linhagens celulares HL-60, HL-60.Bcr- Abl tratadas com TKI (imatinibe, dasatinibe e nilotinibe), KCL22 e LAMA84 resistentes e sensíveis ao MI. Os níveis de expressão dos genes alvo foram correlacionados com o índice de prognóstico de Sokal. Os principais resultados revelaram que há alteração nos genes MST1, MOB1B, MOBKL1B, LATS1, LATS2, TAZ, AURKA e AURKB em pacientes com LMC em relação aos controles. Não houve correlação entre o índice de Sokal e a expressão gênica dos genes da via Hippo, MST1, MOB1B, MOBKL1B, LATS1, LATS2 e TAZ, assim como os genes Aurora quinases A e B. Pacientes com LMC em fases avançadas apresentaram maiores valores de expressão dos genes TAZ e AURKB, comparado aos pacientes na fase crônica. Os pacientes resistentes ao TKI apresentaram as expressões dos genes MST1, TAZ e AURKB, significativamente mais elevadas, comparado aos pacientes sensíveis ao MI. Os resultados dos estudos em linhagens celulares indicaram principalmente que a expressão do gene LATS1 pode ser modulada pela atividade de tirosina quinase Bcr-abl e que o oncogene BCR-ABL1 induz a expressão de AURKA, AURKB, LATS1 e TAZ. Em conjunto os dados obtidos revelam que a alteração da expressão dos genes da família Aurora quinase, A e B, e dos genes que codificam proteínas da via Hippo contribui para a patogênese e progressão da LMC. O desenvolvimento de fármacos e/ou a identificação de marcadores tumorais para a via de sinalização Hippo e família Aurora quinase, podem otimizar o tratamento da LMC, aumentando a susceptibilidade das células leucêmicas a apoptose e levando a um melhor prognóstico da doença / Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm resulting from clonal expansion of myeloid cells positive for the Philadelphia chromosome. The CML pathogenesis is associated with BCR-ABL1 oncogene expression, which encodes the Bcr-Abl protein with a constitutive tyrosine kinase activity, leading to leukemic cell high proliferation and resistance to apoptosis. CML patients are mainly treated with tyrosine kinase inhibitors (TKI), but some of CML patients in chronic phase are resistant and in advanced phases are refractory to TKI. Thus, it is still relevant to elucidate the CML pathogenesis and seek to new therapeutic targets, including the Hippo signaling pathway members and cell cycle regulatory genes such as those encoding the Aurora kinase family. The present study quantified the RNA expression level of genes encoding components from the Hippo cell signaling pathway (MST1, MOB1B, MOBKL1B, LATS1, LATS2, YAP, and TAZ) and Aurora kinase A and B in: 1) CML patients at different stages of the disease, in CML patients resistant or sensitive to imatinib mesylate therapy, healthy individuals and 2) in cell lines HL-60, HL-60.Bcr-Abl treated with TKI (imatinib mesylate, dasatinib and nilotinib), KCL22 and LAMA84 resistant and sensitive to IM. The RNA expression levels of the target genes were also correlated to the CML Sokal\'s prognostic score values. The main results revealed that there are alterations in the genes MST1, MOB1B, MOBKL1B, LATS1, LATS2, TAZ, AURKA and AURKB in patients with CML in relation to the controls. There was no correlation between the Sokal index and the gene expression of the Hippo, MST1, MOB1B, MOBKL1B, LATS1, LATS2 and TAZ genes, as well as the Aurora kinase genes A and B. Patients with advanced phase CML had higher values of expression of the TAZ and AURKB genes, compared to the patients in the chronic phase. Patients resistant to TKI had significantly higher MST1, TAZ and AURKB gene expression compared to MI-sensitive patients. The results of the studies in cell lines indicated primarily that the expression of the LATS1 gene can be modulated by the Bcr-abl tyrosine kinase activity and the BCR-ABL1 oncogene induces the expression of AURKA, AURKB, LATS1 and TAZ. Together, the data show that altered expression of Aurora kinase family genes, A and B, and genes coding for Hippo pathway proteins contribute to the pathogenesis and progression of CML. The development of drugs and/or identification of tumor markers for the Hippo signaling pathway and the Aurora kinase family can optimize CML treatment by enhancing the susceptibility of leukemic cells to apoptosis and leading to a better disease prognosis
30

Arrêt de la prolifération cellulaire pendant le développement embryonnaire : étude transcriptionnelle de gènes suppresseurs de tumeurs au cours de la croissance du système nerveux central chez le poisson médaka Oryzias latipes / Cell proliferation arrest during embryonic development : transcriptionnal study of tumors suppressor genes during central nervous system development in medaka fish Oryzias latipes

Devès, Mathilde 20 September 2012 (has links)
Comment la taille d'un organisme est-elle régulée au cours du développement embryonnaire ? Quels sont les mécanismes génétiques à l'origine de l'arrêt de la prolifération pendant la croissance d'un organisme pluricellulaire ? Afin d'identifier des acteurs de la sortie du cycle cellulaire au cours du développement, mon travail s’est orienté sur l’étude de gènes suppresseurs de tumeurs pendant la croissance du toit optique (TO) du médaka Oryzias latipes. Le TO, structure dorsale du cerveau moyen des Vertébrés, est un modèle particulièrement adapté à l’étude de la régulation de la prolifération. Trois zones de la marge vers le centre du TO sont discernables : une zone périphérique de prolifération, une zone intermédiaire de cellules sortant du cycle cellulaire et une zone centrale de cellules différenciées. Un crible d'expression par hybridation In Situ a été réalisé et a permis d'identifier 28 gènes exprimés dans le TO, suggérant leur implication dans le contrôle de la sortie du cycle cellulaire au cours du développement. Dans le but de caractériser in vivo la fonction de gènes issus de ce crible, le gène BTG1 (B-cell Translocation Gene 1) et les membres de sa famille, ont été étudiés au cours du développement du médaka. J’ai mené des expériences fonctionnelles sur BTG1, permettant de mettre en évidence son rôle central pour la morphogenèse du système nerveux central. De plus, une autre partie de mon travail s’est penchée sur l’étude de l’expression des membres de la voie de signalisation Hippo, bien connue et caractérisée chez la drosophile et les Mammifères pour son rôle dans le contrôle de la taille des organes via une régulation de l’arrêt de la prolifération. A l’issu de notre travail, une fonction de la voie de signalisation Hippo dans la formation du TO et des somites a pu être mise en évidence au cours du développement du médaka. / How is an organisms’ size regulated during embryonic development? What are the genetic mechanisms that control the proliferation arrest during multicellular organisms growth? In order to identify a cell cycle exit developmental actor genes, I have analysed the role of tumor suppressor genes (TSGs) in the optic tectum (OT) of the medaka Oryzias latipes. This structure is particularly suited for this kind of studies because, during its morphogenesis, there is a strict correlation between the position of a cell and its degree of differentiation. 3 zones can be easily distinguished from the border to the center: a marginal zone made of proliferative cells, an intermediate zone in which cells exit the cycle, and a central zone made of postmitotic cells. Using this criterium, I have performed an in situ hybridization expression screen on 150 TSGs on medaka embryos. The expression patterns of 28 TSGs in the OT suggest their implication in the OT proliferation arrest mechanisms. I focused my study on the BTG1 gene, implicated in many cancers, and for which few developmental data are available. A functional analysis on developing medaka embryos has been performed and permitted to highlight the essential role of BTG1 in central nervous system morphogenesis. Furthermore, I performed an expression study on Hippo signalling pathway components. Hippo pathway is well caracterised for its organ size control function in drosophila and Mammals. Our results show that this pathway could act in OT formation and somitogenesis in medaka fish.

Page generated in 0.0342 seconds