18 research outputs found

    METTL13 methylation of eEF1A increases translational output to promote tumorigenesis

    Full text link
    Increased protein synthesis plays an etiologic role in diverse cancers. Here, we demonstrate that METTL13 (methyltransferase-like 13) dimethylation of eEF1A (eukaryotic elongation factor 1A) lysine 55 (eEF1AK55me2) is utilized by Ras-driven cancers to increase translational output and promote tumorigenesis in vivo. METTL13-catalyzed eEF1A methylation increases eEF1A's intrinsic GTPase activity in vitro and protein production in cells. METTL13 and eEF1AK55me2 levels are upregulated in cancer and negatively correlate with pancreatic and lung cancer patient survival. METTL13 deletion and eEF1AK55me2 loss dramatically reduce Ras-driven neoplastic growth in mouse models and in patient-derived xenografts (PDXs) from primary pancreatic and lung tumors. Finally, METTL13 depletion renders PDX tumors hypersensitive to drugs that target growth-signaling pathways. Together, our work uncovers a mechanism by which lethal cancers become dependent on the METTL13-eEF1AK55me2 axis to meet their elevated protein synthesis requirement and suggests that METTL13 inhibition may constitute a targetable vulnerability of tumors driven by aberrant Ras signaling.We thank Pal Falnes, Jerry Pelletier, and Julien Sage for helpful discussion, Lauren Brown and William Devine for SDS-1-021, and members of the Gozani and Mazur laboratories for critical reading of the manuscript. This work was supported in part by grants from the NIH to S.M.C. (K99CA190803), M.P.K. (5K08CA218690-02), J.A.P. (R35GM118173), M.C.B. (1DP2HD084069-01), J.S. (1R35GM119721), I.T. (R01CA202021), P.K.M. (R00CA197816, P50CA070907, and P30CA016672), and O.G. (R01GM079641). J.E.E. received support from Stanford ChEM-H, and A.M. was supported by the MD Anderson Moonshot Program. I.T. is a Junior 2 Research Scholar of the Fonds de Recherche du Quebec - Sante (FRQ-S). P.K.M. is supported by the Neuroendocrine Tumor Research Foundation and American Association for Cancer Research and is the Andrew Sabin Family Foundation Scientist and CPRIT scholar (RR160078). S.H. is supported by a Deutsche Forschungsgemeinschaft Postdoctoral Fellowship. J.W.F. is supported by 5T32GM007276. (K99CA190803 - NIH; 5K08CA218690-02 - NIH; R35GM118173 - NIH; 1DP2HD084069-01 - NIH; 1R35GM119721 - NIH; R01CA202021 - NIH; R00CA197816 - NIH; P50CA070907 - NIH; P30CA016672 - NIH; R01GM079641 - NIH; Stanford ChEM-H; MD Anderson Moonshot Program; Neuroendocrine Tumor Research Foundation; American Association for Cancer Research; Deutsche Forschungsgemeinschaft Postdoctoral Fellowship; 5T32GM007276)Supporting documentationAccepted manuscrip

    Mécanismes différentiels de répression transcriptionnelle des gènes cibles de HIC1

    No full text
    HIC1 (Hypermethylated in Cancer 1) is a transcriptional repressor encoded by a tumor suppressor gene localised in 17p13.3. In many human cancers this region is lost or inactivated by hypermethylation; and heterozygous mice Hic+/- develop spontaneous tumors with a higher incidence than control mice.HIC1 is implicated in complex regulatory loops together with p53, the class III deacetylase SIRT1 and a cell cycle control protein, E2F1.During the DNA damage response, HIC1 represses SIRT1 transcription, leading to the accumulation of active acetylated p53 and thus to apoptosis and cell cycle arrest. In addition HIC1 is a direct target gene activated by p53. Metabolism also regulates this pathway since the transcriptional repression of SIRT1 by HIC1 required the corepressor CtBP, which is regulated by the NADH/NAD+ balance.Another loop connected to the first in the DNA damage response implicates E2F1 that activates HIC1 transcription. This is also a feedback regulatory pathway since HIC1 represses E2F1 transcription in at least G0 quiescent cells.In this study, we have investigated the transcriptional repression mechanisms brought about by HIC1 on known as well as newly identified target genes.We have identified MTA1, a member of the NuRD complexes, as a new HIC1's corepressor whose recruitment is controlled by Lysine 314 SUMOylation/Acetylation switch. In agreement with HIC1's roles in growth control, this HIC1-MTA1 complex is bound to two new target genes promoters p57KIP2 and Cyclin D1, in quiescent cells, as well as on a new conserved site of the SIRT1 promoter.NuRD complexes regulate the majority of HIC1's targets genes whereas CtBP only repressed SIRT1 and the recently identified gene, CXCR7.HIC1 also interacts with a subset of SWI/SNF complexes containing the BRG1 ATPase and the specific unit ARID1A. This interaction is required to repress E2F1, but not SIRT1, in quiescent fibroblasts.Our results suggest that the HIC1's transcriptional repression mechanisms are tightly regulated and specific according to targets genes and cells status.HIC1 (Hypermethylated in Cancer 1) est un répresseur transcriptionnel codé par un gène suppresseur de tumeurs localisé en 17p13.3. Cette région est perdue ou inactivée par hyperméthylation dans de nombreux cancers humains ; et les souris hétérozygotes Hic+/- développent des tumeurs spontanées avec une incidence beaucoup plus élevée que les souris contrôle.Cette protéine est impliquée dans des boucles de régulation complexes impliquant p53, la désacétylase de classe III SIRT1 ainsi qu'une des protéines de contrôle du cycle cellulaire, E2F1.En réponse aux dommages à l'ADN, HIC1 réprime SIRT1, ce qui a pour conséquence l'augmentation du taux de p53 acétylée active. Ceci conduit à l'apoptose et à l'arrêt du cycle cellulaire. HIC1 étant lui-même activé par p53, cette boucle peut s'auto entretenir. Cette voie est également régulée par le métabolisme puisque la répression de SIRT1 par HIC1 est due, notamment, au corépresseur CtBP, lui-même régulé par la balance NADH/NAD+.D'autre part, et de manière intrinsèquement liée, cette même réponse aux dommages à l'ADN induit l'expression de HIC1 par E2F1. Ceci mène à une seconde boucle de régulation puisque HIC1 réprime E2F1, notamment lors de la phase de quiescence G0.Cette présente étude porte sur les différents mécanismes de répression transcriptionnelle mis en place par HIC1, sur ses gènes cibles déjà connus et nouvellement identifiés.Nous avons pu identifier un nouveau corépresseur de HIC1, MTA1, un membre du complexe NuRD, dont le recrutement est contrôlé par la compétition SUMOylation/Acétylation de la Lysine 314 de HIC1. De manière cohérente avec le rôle de HIC1 dans le contrôle de la croissance, le complexe HIC1-MTA1 est lié au promoteur de nouveaux gènes cibles, p57KIP2 et Cycline D1, dans des cellules quiescentes, ainsi qu'à un site nouvellement identifié au sein du promoteur de SIRT1.Tandis que le complexe NuRD apparaît réguler une majorité des gènes cibles de HIC1 connus à ce jour, ce n'est pas le cas pour CtBP, qui régulerait SIRT1 et un gène identifié récemment, CXCR7.De plus, HIC1 interagit avec le complexe SWI/SNF composé de l'ATPase BRG1 et de la sous-unité appartenant aux complexes répresseurs ARID1A, et ce pour réprimer E2F1, mais pas SIRT1, au sein de cellules primaires quiescentes.Ces résultats suggèrent la mise en place par HIC1 de mécanismes de répression transcriptionnelle complexes et finement régulés en fonction du type de gènes cibles et de l'état de la cellule

    Hypermethylated in cancer 1 (HIC1) recruits polycomb repressive complex 2 (PRC2) to a subset of its target genes through interaction with human polycomb-like (hPCL) proteins

    No full text
    HIC1 (hypermethylated in cancer 1) is a tumor suppressor gene epigenetically silenced or deleted in many human cancers. HIC1 is involved in regulatory loops modulating p53- and E2F1-dependent cell survival, growth control, and stress responses. HIC1 is also essential for normal development because Hic1-deficient mice die perinatally and exhibit gross developmental defects throughout the second half of development. HIC1 encodes a transcriptional repressor with five C(2)H(2) zinc fingers mediating sequence-specific DNA binding and two repression domains: an N-terminal BTB/POZ domain and a central region recruiting CtBP and NuRD complexes. By yeast two-hybrid screening, we identified the Polycomb-like protein hPCL3 as a novel co-repressor for HIC1. Using multiple biochemical strategies, we demonstrated that HIC1 interacts with hPCL3 and its paralog PHF1 to form a stable complex with the PRC2 members EZH2, EED, and Suz12. Confirming the implication of HIC1 in Polycomb recruitment, we showed that HIC1 shares some of its target genes with PRC2, including ATOH1. Depletion of HIC1 by siRNA interference leads to a partial displacement of EZH2 from the ATOH1 promoter. Furthermore, in vivo, ATOH1 repression by HIC1 is associated with Polycomb activity during mouse cerebellar development. Thus, our results identify HIC1 as the first transcription factor in mammals able to recruit PRC2 to some target promoters through its interaction with Polycomb-like proteins
    corecore