• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 3
  • 2
  • Tagged with
  • 5
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 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

Ribozomálny proteín Rpl22 reguluje zostrih svojich vlastných transcriptov / Ribosomal protein Rpl22 regulates the splicing of its own transcripts

Nemčko, Filip January 2018 (has links)
Saccharomyces cerevisiae is an intron-poor organism with introns present in only 5% of its genes. The most prominent group of intron-containing genes are ribosomal protein (RP) genes. They are highly expressed and most of them are present as two paralogs. Parenteau et al. described the existence of intron- dependent intergenic regulatory circuits controlling expression ratios of RP paralogs. In this project, we did not confirm the regulation in 6 out of 7 tested regulatory circuits. We validated the regulation between RPL22 paralogs. We further showed that Rpl22 protein blocks the pre-mRNA splicing of both paralogs, with RPL22B paralog being more sensitive. Rpl22 protein binds to the stem-loop of RPL22B intron - disruption of the binding domain of Rpl22 proteins leads to loss of interaction. Moreover, the regulation seems to be working the same way in yeast Kluyveromyces lactis, which has only a single RPL22 copy. Overall, these results lead to better understanding of intergenic regulation, which adjusts the expression ratio between functionally different RPL22 paralogs. Key words introns, ribosomal protein genes, Rpl22, RPL22 paralogs, pre-mRNA splicing, Saccharomyces cerevisiae
2

Ribozomálny proteín Rpl22 reguluje zostrih svojich vlastných transcriptov / Ribosomal protein Rpl22 regulates the splicing of its own transcripts

Nemčko, Filip January 2018 (has links)
Saccharomyces cerevisiae is an intron-poor organism with introns present in only 5% of its genes. The most prominent group of intron-containing genes are ribosomal protein (RP) genes. They are highly expressed and most of them are present as two paralogs. Parenteau et al. described the existence of intron- dependent intergenic regulatory circuits controlling expression ratios of RP paralogs. In this project, we did not confirm the regulation in 6 out of 7 tested regulatory circuits. We validated the regulation between RPL22 paralogs. We further showed that Rpl22 protein blocks the pre-mRNA splicing of both paralogs, with RPL22B paralog being more sensitive. Rpl22 protein binds to the stem-loop of RPL22B intron - disruption of the binding domain of Rpl22 proteins leads to loss of interaction. Moreover, the regulation seems to be working the same way in yeast Kluyveromyces lactis, which has only a single RPL22 copy. Overall, these results lead to better understanding of intergenic regulation, which adjusts the expression ratio between functionally different RPL22 paralogs. Key words introns, ribosomal protein genes, Rpl22, RPL22 paralogs, pre-mRNA splicing, Saccharomyces cerevisiae
3

EXTRARIBOSOMAL REGULATION OF MYELOID LEUKEMOGENESIS BY RPL22

Harris, Bryan January 2019 (has links)
Mutations and deletions in ribosomal proteins are associated with a group of diseases termed ribosomopathies. Collectively, these diseases are characterized by ineffective hematopoiesis, bone marrow failure, and an increased risk of developing myelodysplastic syndrome (MDS) and subsequently acute myeloid leukemia (AML). This observation highlights the role of dysregulation of this class of proteins in the development and progression of myeloid neoplasms. Analysis of gene expression in CD34+ hematopoietic stem cells (HSC) from 183 MDS patients demonstrated that ribosomal protein L22 (Rpl22) expression exhibited a greater reduction than any other ribosomal protein gene in MDS. Interestingly, we observed that AML patients with lower expression of Rpl22 had a significant reduction in their survival (TCGA cohort, N=200, Log Rank P value<0.05). To assess the mechanism of reduced expression, we developed a FISH probe complementary to the RPL22 locus and assessed for deletion of this locus in an independent set of 104 MDS/AML bone marrow samples. Strikingly, we found that RPL22 deletion was enriched in high-risk MDS and secondary AML cases. We, therefore, sought to investigate whether reduced Rpl22 expression played a causal role in leukemogenesis. Using Rpl22-/- mice, we found that Rpl22-deficiency resulted in a constellation of phenotypes resembling MDS. Indeed, Rpl22-deficiency caused a macrocytic reduction in red blood cells, dysplasia in the bone marrow, and an expansion of the early hematopoietic stem and progenitor compartment (HSPC). Since MDS has been described as a disease originating from the stem cell compartment, we next sought to determine if the hematopoietic defects were cell autonomous and resident in Rpl22-/- HSC. Competitive transplantation revealed that Rpl22-/- HSC exhibited pre-leukemic characteristics including effective engraftment, but a failure to give rise to downstream mature blood cell lineages. Importantly, there was a strong myeloid bias in those downstream progeny derived form Rpl22-/- HSC. To determine how Rpl22-deficiency increased the causes these deficiencies in HSC, we performed whole transcriptome analysis on Rpl22-/- HSC. Interestingly, alterations in genes associated with both ribosomal proteins and mitochondrial components were observed. We found that protein synthesis was unchanged in Rpl22-deficient HSCs, sharply contrasting the reductions in global protein synthesis that usually accompany ribosomal protein insufficiency. Consequently, we shifted our focus to the dysregulated mitochondrial genes, which were linked to the processes of oxidative phosphorylation and fatty acid metabolism. We observed that oxidative phosphorylation was decreased in Rpl22-deficient HSCs while fatty acid oxidation was increased. Increased fatty acid oxidation is associated with maintenance of the hematopoietic stem cells. Interestingly, inhibiting fatty acid oxidation mitigated this attribute in Rpl22 deficient HSCs. Because Rpl22 is an RNA-binding protein, we asked if Rpl22 was regulating fatty acid oxidation by directly binding mRNAs encoding regulators of fatty acid oxidation. We found that Rpl22 is able to directly bind the coding region of an upstream regulator of fatty acid oxidation, Alox12. Thus, we hypothesized that Rpl22-deficiency increased fatty acid oxidation through increased expression of Alox12. Consistent with this hypothesis, knockdown of Alox12 impaired the function of Rpl22 deficient HSC. Because the increased fatty acid oxidation promotes self-renewal of Rpl22-deficient HSC and blocks their differentiation, we also hypothesized that this would predispose them to leukemogenesis. We examined the potential for Rpl22-deficient HSPC to be transformed upon ectopic expression of the MLL-AF9 oncogenic fusion. Indeed, Rpl22 deficiency increased predisposition to transformation both in vitro and in vivo, in MLL-AF9 knockin mice. Furthermore, Rpl22 deficient leukemias were preferentially sensitive to pharmacologic inhibition of fatty acid oxidation or Alox12 knockdown, indicating that leukemia cell survival was also dependent upon fatty acid oxidation. Taken together, these findings indicate that Rpl22-insufficiency predisposes HSPC to leukemic transformation and aggressive growth by regulating mitochondrial function, providing an explanation for the reduced survival observed in Rpl22-low AML patients. We also sought to determine how Rpl22 may be contributing to another subset of AML known as Therapy-related AML. Most commonly, these patients develop AML after previously being treated with an alkylating chemotherapeutic drug. Interestingly, we found that Rpl22-deficient HSPC are resistant to treatment with these agents, despite having evidence of DNA damage. The ultimate consequence of the insensitivity of Rpl22-deficient HSPC to alkylating agents was that mice given serial doses of cyclophosphamide exhibited an increased incidence of leukemic-like changes. This chemo-resistant phenotype in Rpl22-/- cells was related to increased expression of the DNA repair protein MGMT. Inhibition of this protein abrogated the ability of these cells to survive following treatment with cyclophosphamide. Ultimately, this study implicates Rpl22 as a regulator of alkylating DNA damage repair and suggests that both patients with hematologic or solid cancers that express reduced levels of Rpl22 are at increased risk for development of therapy related AML is they are treated with alkylating agents. / Cancer Biology & Genetics
4

Rôle du ribosome dans la sénescence

Del Toro Del Toro, Neylen 12 1900 (has links)
La sénescence est considérée comme un mécanisme de suppression tumorale puisque les cellules potentiellement dangereuses, activent leurs protéines de sauvegarde pour arrêter leur prolifération. Les protéines de sauvegarde telles que RB et p53 sont activées suite à différents stress comme des dommages à l’ADN, le raccourcissement des télomères ou l’induction oncogénique. Les cellules sénescentes restent métaboliquement actives, subissent des modifications dans leur expression génique, et sécrètent des cytokines et des chimiokines qui ont des effets paracrines pro-oncogéniques, mais peuvent également contribuer à la stabilité de l’arrêt du cycle cellulaire dans la sénescence de façon autocrine. Une des particularités du phénotype sénescent est la dégradation sélective des protéines dépendante de l’ubiquitination et du protéasome. Parmi les cibles de dégradation se trouvent des protéines impliquées dans la biogenèse du ribosome, ainsi que celles d’autres voies cellulaires requises pour la croissance de cellules cancéreuses. Ceci est lié à un stress nucléolaire qui affecte la biogenèse du ribosome, menant à l’accumulation, dans le nucléoplasme ou le nucléole, de protéines ribosomiques. Ce comportement suggère que les ribosomes des cellules sénescentes seraient structurellement différents. Par conséquent, ceci pourrait entrainer des effets sur leurs capacités à réguler l’initiation, l’élongation et/ou la terminaison de la traduction des ARN messagers (ARNm). Par ailleurs, la déplétion de certaines protéines impliquées dans la ribogenèse, ainsi que la surexpression de protéines ribosomiques telles que RPS14/uS11 amènent à la sénescence. Malgré le stress nucléolaire et les défauts de ribogenèse associés à la sénescence, les cellules sénescentes présentent des niveaux de translecture du codon d’arrêt très diminué, suggérant l’existence de défauts de production de protéines allongées en C-terminal. Nous émettons l’hypothèse que les défauts de la ribogenèse affecteraient la fonction des protéines ribosomiques et des ribosomes. Cette perturbation aurait un impact sur le rôle de suppresseur tumoral de la sénescence. Le premier objectif de cette thèse consiste à démontrer le rôle de RPL22/eL22 en tant que régulateur du cycle cellulaire et inducteur de la sénescence. Le deuxième but est de démontrer que, malgré la perturbation nucléolaire, les ribosomes des fibroblastes sénescents reconnaissent les codons d’arrêt de façon plus efficace que les ribosomes des cellules transformées, ou des cellules normales en prolifération. Nous avons démontré que le phénotype de sénescence peut être induit quand l’expression de RPL22/eL22 est augmentée. RPL22/eL22 s’accumule principalement dans le nucléole, de manière différente de RPS14/uS11, dont l’accumulation est nucléoplasmique. En effectuant des essais kinases in vitro, nous avons montré que RPL22/eL22, tout comme RPS14/uS11, peuvent interagir et inhiber le complexe CDK4-Cycline D1 afin d’activer la voie de RB et établir l’arrêt du cycle cellulaire et la sénescence. Afin de démontrer la fidélité de la terminaison de la traduction dans les cellules sénescentes, nous avons utilisé un système de rapporteurs de luciférases, pour détecter les erreurs de translecture ainsi que pour avoir un contrôle interne du système. L’inactivation de la voie du suppresseur tumoral RB par surexpression de CDK4 ou de l’oncoprotéine virale E7, nous a permis d’observer l’augmentation de la translecture dans les cellules sénescentes. Tandis que l’activation de la voie de suppression tumorale RB, à l’aide du suppresseur de tumeur PML, de la surexpression de RPL22/eL22 et de RPS14/uS11, ainsi que de l’utilisation de Palbociclib (PD-0332991), un inhibiteur des kinases CDK4/6, a montré une réduction des erreurs de translecture. Ces résultats indiquent une nouvelle fonction des protéines du ribosome en tant que suppresseurs de tumeur, permettant d’inhiber les erreurs de translecture du codon d’arrêt de façon dépendante de la voie de RB. Ces travaux suggèrent que de petites molécules ou peptides pourraient simuler les fonctions inhibitrices de ces protéines ribosomiques afin de traiter certains cancers où la voie de RB est activable. / Senescence is considered a mechanism for tumor suppression since potentially dangerous cells activate their protective proteins to stop their proliferation. Safeguard proteins such as RB and p53 are activated as a result of stress such as DNA damage, telomere shortening or oncogenic induction. Senescent cells are metabolically active, they undergo changes in their gene expression and secrete cytokines and chemokines with pro-oncogenic paracrine effects, but which can also contribute to the stability of the senescent cell cycle arrest in an autocrine way. One of the peculiarities of the senescent phenotype is the selective ubiquitination and proteasome dependent-degradation of proteins involved in ribosome biogenesis and other cellular pathways required for cancer cell growth, leading to the accumulation, in the nucleoplasm or nucleolus, of ribosomal proteins. This behavior suggests that the ribosomes of senescent cells are structurally different. Therefore, this could have effects on their ability to regulate the initiation, elongation and/or translation termination of messenger RNAs (mRNAs). Moreover, the depletion of some proteins involved in ribogenesis, as well as the overexpression of ribosomal proteins such as RPS14/uS11 lead to senescence. Despite nucleolar stress and ribogenesis defects associated to senescence, global translation does not seem to be affected in senescence. Strikingly, senescent cells have reduced translational readthrough suggesting that they have defects in the production of C-terminal extended proteins. We hypothesize that defects in ribogenesis would affect the function of ribosomal proteins and ribosomes influencing the tumor suppressor role of senescence. The first aim of this thesis is to demonstrate the role of RPL22/eL22 as a regulator of the cell cycle and senescence inducer. The second aim of this thesis is to demonstrate that, despite the nucleolar disruption, the ribosomes of senescent fibroblasts recognize stop codons more efficiently than ribosomes from transformed cells, but also than ribosomes from proliferating normal cells. We found that the senescent phenotype can be induced by enhancing the expression of RPL22/eL22. RPL22/eL22 accumulates mainly in the nucleolus, unlike RPS14/uS11, whose accumulation is nucleoplasmic. By performing an in vitro kinase assay, we showed that RPL22/eL22, just like RPS14/uS11, can interact and inhibit the CDK4-Cyclin D1 complex in order to activate the RB pathway and establish cellular arrest and senescence. To assess translation termination accuracy in senescent cells, we used a system of luciferase reporters to measure the fidelity of translation termination. Inactivation of the RB tumor suppressor pathway using CDK4 or the viral oncoprotein E7 also increased readthrough in senescent cells while overexpression of PML, a tumor suppressor that activates the RB pathway, overexpression of RPL22/eL22 and RPS14/uS11, as well as the use of Palbociclib (PD-0332991), a CDK4/6 inhibitor, reduce readthrough errors. These results indicate a novel function of ribosomal proteins as tumor suppressors, making it possible to inhibit translational readthrough errors, in a RB-dependent pathway. This work suggests that small molecules or peptides could mimic the inhibitory functions of these ribosomal proteins in order to treat cancers where the RB pathway is activatable.
5

Mécanismes de régulation post-traductionnelle de la sénescence cellulaire et leurs impacts sur la suppression tumorale

Fernandez Ruiz, Ana 07 1900 (has links)
La sénescence est un processus caractérisé par un arrêt stable du cycle cellulaire. Ce mécanisme peut être induit en réponse à de nombreux stress, comme l’activation d’un oncogène, le raccourcissement des télomères ou bien le traitement avec des composés génotoxiques. Cette réponse cellulaire est considérée comme une barrière antitumorale limitant la prolifération des cellules exposées au risque de transformation. La mise en place de la sénescence dépend de profonds changements au niveau moléculaire, dont l’activation d’un programme de dégradation sélective des protéines. Cette dégradation de protéines associée à la sénescence (SAPD) peut expliquer plusieurs caractéristiques des cellules sénescentes, notamment la présence de défauts dans la voie de synthèse des ribosomes (SARD). Ces derniers sont liés à un stress nucléolaire qui mène à l’accumulation de certaines protéines ribosomiques dans le noyau, où elles peuvent effectuer des fonctions indépendantes de leur rôle structurale dans les ribosomes. Parmi ces protéines ribosomiques, RPS14/uS11 peut s’accumuler dans le nucléoplasme et réguler le cycle cellulaire en inhibant CDK4. Ces mécanismes de régulation post-traductionnelle -le SAPD ainsi que les conséquences des SARD- contribuent de manière importante au phénotype sénescent. Nous avons émis l’hypothèse que la caractérisation des effecteurs dans ces voies pourrait mener à l’identification de nouvelles protéines importantes pour la sénescence et la suppression tumorale. Dans un premier temps, nous avons évalué le rôle de la protéine ribosomique RPL22/eL22 dans le cycle cellulaire et la sénescence. Tout comme RPS14, RPL22 a été identifié dans l’analyse de l’interactome de CDK4 lors de la sénescence induite par la perte du facteur de la ribogenèse RSL1D1. Nous avons pensé que RPL22 pourrait agir de manière similaire à RPS14 et ainsi effectuer des fonctions extra-ribosomiques impliquées dans la régulation du cycle cellulaire. Dans le premier article présenté dans cette thèse, nous montrons que la surexpression de RPL22 dans des fibroblastes humains induit un phénotype sénescent et que RPL22 peut lier et inhiber CDK4 afin d’activer la voie de RB. Ensemble, ces données indiquent un rôle suppressif de RPL22 dans le cycle cellulaire. En second lieu, nous nous sommes penchés sur la caractérisation des effecteurs du programme de dégradation sélective de protéines associé à la sénescence. Ce programme est mené à terme par le système ubiquitine-protéasome, un mécanisme finement régulé par différents types de protéines. Parmi celles-ci, les E3 ubiquitine ligases définissent la spécificité de ce système en interagissant avec les substrats à dégrader. Nous avons donc pensé que certaines E3 ubiquitine ligases spécifiques pourraient être importantes pour le mécanisme de dégradation protéique associé à la sénescence. Afin d’identifier celles-ci, nous avons effectué un criblage de shARN ciblant des gènes d’E3 ubiquitine ligases dans le contexte de la sénescence induite par les oncogènes. Ceci a mené à l’identification d’ASB14 comme un acteur important de la sénescence. Dans le deuxième article de cette thèse, nous montrons que la perte d’ASB14 produit un contournement de la sénescence induite par l’oncogène RAS dans plusieurs modèles cellulaires. ASB14 est une protéine peu caractérisée et nous avons généré des anticorps afin d’analyser son expression. Nous montrons ensuite qu’ASB14 s’exprime fortement dans le pancréas sain, tandis que ses niveaux diminuent dans les tumeurs pancréatiques. Enfin, nous avons identifié les partenaires d’interaction d’ASB14 dans le contexte de la sénescence induite par l’oncogène RAS. Globalement, les travaux présentés dans cette thèse nous ont permis d’identifier deux nouvelles protéines impliquées dans la sénescence cellulaire : la protéine ribosomique RPL22 et l’E3 ubiquitine ligase ASB14. Ces deux protéines contribuent à la régulation post-traductionnelle du phénotype sénescent. D’un côté, RPL22 peut inhiber l’activité de CDK4 afin d’activer la voie de RB et ainsi réguler le cycle cellulaire. D’une autre part, ASB14 est importante pour le maintien du phénotype sénescent et semble avoir un rôle dans la suppression tumorale du pancréas. Nos résultats suggèrent que RPL22 et ASB14 sont importants pour la sénescence et la suppression tumorale. / Cellular senescence is characterized by a stable cell cycle arrest. This process can be induced by a variety of cellular stresses, including oncogene activation, telomere shortening and genotoxic treatments. In fact, senescence is considered an antitumor barrier that prevents cellular transformation. Senescence is associated with widespread molecular changes, including the activation of a selective protein degradation program. This senescence-associated protein degradation (SAPD) could regulate some senescence-associated phenotypes, including the senescence-associated ribosome biogenesis defects (SARD). Senescence-associated ribosome biogenesis defects are linked to a nuclear accumulation of some ribosomal proteins such as RPS14/uS11 capable of carrying out extra-ribosomal functions. In particular, RPS14 can inhibit CDK4 and mediate senescence. Thus, we hypothesize that the proteins implicated in these pathways -SAPD and SARD- could be important for senescence and tumor suppression. First, we evaluated the ability of the ribosomal protein L22 (RPL22/eL22) to regulate cellular senescence and cell cycle progression. RPL22, as RPS14, was identified as a binding partner for CDK4 in senescent cells induced by depleting the ribosome biogenesis factor RSL1D1. Hence, we though that RPL22 could act in a manner similar to RPS14. In chapter two, we show that RPL22 overexpression induces a senescent phenotype in human fibroblasts. In addition, we show that RPL22 can interact with CDK4 inhibiting its activity and stimulating the RB tumor suppressor pathway. Taken together, these results indicate a suppressive role of RPL22 in cell cycle progression. Next, we focused on the characterization of SAPD effectors. This mechanism is mediated by the ubiquitin-proteasome system which is tightly regulated by E3 ubiquitin ligases. Thus, we thought that specific E3 ubiquitin ligases could be important for SAPD and for senescence. In order to discover E3 ubiquitin ligases that contribute to senescence, we performed an unbiased screening using shRNA libraries in Ras-induced senescent cells. This led to the identification of ASB14 as an important mediator of senescence. In chapter three, we show that ASB14 depletion leads to a bypass of Ras-induced senescence. ASB14 is a poorly characterized E3 ligase, and we generated antibodies in order to analyze its expression levels. We show that ASB14 is highly expressed in the normal pancreas whereas its expression is reduced in pancreatic cancer tissues. Finally, we uncovered the interactome of ASB14 in Ras-induced senescent cells. Overall, we have discovered two new senescence mediators: ribosomal protein L22 and E3 ubiquitin ligase ASB14. These proteins are implicated in the post-translational regulation of the senescent phenotype. RPL22 acts as a CDK4 inhibitor to activate RB pathway and regulate cell cycle arrest and ASB14 is an important mediator of senescence maintenance. Taken together, our results suggest that RPL22 and ASB14 are important for cellular senescence and tumor suppression.

Page generated in 0.0874 seconds