Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37398006

ABSTRACT

Epigenetic modifications that arise during plant and animal development, such as DNA and histone modification, are mostly reset during gamete formation, but some are inherited from the germline including those marking imprinted genes1. Small RNAs guide these epigenetic modifications, and some are also inherited by the next generation2,3. In C. elegans, these inherited small RNAs have poly (UG) tails4, but how inherited small RNAs are distinguished in other animals and plants is unknown. Pseudouridine (Ψ) is the most abundant RNA modification but has not been explored in small RNAs. Here, we develop novel assays to detect Ψ in short RNA sequences, demonstrating its presence in mouse and Arabidopsis microRNAs and their precursors. We also detect substantial enrichment in germline small RNAs, namely epigenetically activated siRNAs (easiRNAs) in Arabidopsis pollen, and piwi-interacting piRNAs in mouse testis. In pollen, pseudouridylated easiRNAs are localized to sperm cells, and we found that PAUSED/HEN5 (PSD), the plant homolog of Exportin-t, interacts genetically with Ψ and is required for transport of easiRNAs into sperm cells from the vegetative nucleus. We further show that Exportin-t is required for the triploid block: chromosome dosage-dependent seed lethality that is epigenetically inherited from pollen. Thus, Ψ has a conserved role in marking inherited small RNAs in the germline.

2.
Commun Biol ; 5(1): 868, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36008532

ABSTRACT

RNA methylation plays an important role in functional regulation of RNAs, and has thus attracted an increasing interest in biology and drug discovery. Here, we collected and collated transcriptomic, proteomic, structural and physical interaction data from the Harmonizome database, and applied supervised machine learning to predict novel genes associated with RNA methylation pathways in human. We selected five types of classifiers, which we trained and evaluated using cross-validation on multiple training sets. The best models reached 88% accuracy based on cross-validation, and an average 91% accuracy on the test set. Using protein-protein interaction data, we propose six molecular sub-networks linking model predictions to previously known RNA methylation genes, with roles in mRNA methylation, tRNA processing, rRNA processing, but also protein and chromatin modifications. Our study exemplifies how access to large omics datasets joined by machine learning methods can be used to predict gene function.


Subject(s)
Machine Learning , Proteomics , Humans , Methylation , RNA , Supervised Machine Learning
3.
Gut ; 2022 01 10.
Article in English | MEDLINE | ID: mdl-35012996

ABSTRACT

OBJECTIVE: Intratumor heterogeneity drives cancer progression and therapy resistance. However, it has yet to be determined whether and how subpopulations of cancer cells interact and how this interaction affects the tumour. DESIGN: We have studied the spontaneous flow of extracellular vesicles (EVs) between subpopulations of cancer cells: cancer stem cells (CSC) and non-stem cancer cells (NSCC). To determine the biological significance of the most frequent communication route, we used pancreatic ductal adenocarcinoma (PDAC) orthotopic models, patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMMs). RESULTS: We demonstrate that PDAC tumours establish an organised communication network between subpopulations of cancer cells using EVs called the EVNet). The EVNet is plastic and reshapes in response to its environment. Communication within the EVNet occurs preferentially from CSC to NSCC. Inhibition of this communication route by impairing Rab27a function in orthotopic xenographs, GEMMs and PDXs is sufficient to hamper tumour growth and phenocopies the inhibition of communication in the whole tumour. Mechanistically, we provide evidence that CSC EVs use agrin protein to promote Yes1 associated transcriptional regulator (YAP) activation via LDL receptor related protein 4 (LRP-4). Ex vivo treatment of PDXs with antiagrin significantly impairs proliferation and decreases the levels of activated YAP.Patients with high levels of agrin and low inactive YAP show worse disease-free survival. In addition, patients with a higher number of circulating agrin+ EVs show a significant increased risk of disease progression. CONCLUSION: PDAC tumours establish a cooperation network mediated by EVs that is led by CSC and agrin, which allows tumours to adapt and thrive. Targeting agrin could make targeted therapy possible for patients with PDAC and has a significant impact on CSC that feeds the tumour and is at the centre of therapy resistance.

4.
Nat Commun ; 12(1): 7198, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893601

ABSTRACT

RNA molecules undergo a vast array of chemical post-transcriptional modifications (PTMs) that can affect their structure and interaction properties. In recent years, a growing number of PTMs have been successfully mapped to the transcriptome using experimental approaches relying on high-throughput sequencing. Oxford Nanopore direct-RNA sequencing has been shown to be sensitive to RNA modifications. We developed and validated Nanocompore, a robust analytical framework that identifies modifications from these data. Our strategy compares an RNA sample of interest against a non-modified control sample, not requiring a training set and allowing the use of replicates. We show that Nanocompore can detect different RNA modifications with position accuracy in vitro, and we apply it to profile m6A in vivo in yeast and human RNAs, as well as in targeted non-coding RNAs. We confirm our results with orthogonal methods and provide novel insights on the co-occurrence of multiple modified residues on individual RNA molecules.


Subject(s)
Nanopore Sequencing/methods , Nanopores , RNA/metabolism , Sequence Analysis, RNA/methods , Base Sequence , Computational Biology , Gene Expression Profiling , Genetic Techniques , High-Throughput Nucleotide Sequencing , Humans , RNA/isolation & purification , RNA Processing, Post-Transcriptional , Software , Transcriptome
5.
Sci Adv ; 7(27)2021 06.
Article in English | MEDLINE | ID: mdl-34193418

ABSTRACT

The global outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) necessitates the rapid development of new therapies against coronavirus disease 2019 (COVID-19) infection. Here, we present the identification of 200 approved drugs, appropriate for repurposing against COVID-19. We constructed a SARS-CoV-2-induced protein network, based on disease signatures defined by COVID-19 multiomics datasets, and cross-examined these pathways against approved drugs. This analysis identified 200 drugs predicted to target SARS-CoV-2-induced pathways, 40 of which are already in COVID-19 clinical trials, testifying to the validity of the approach. Using artificial neural network analysis, we classified these 200 drugs into nine distinct pathways, within two overarching mechanisms of action (MoAs): viral replication (126) and immune response (74). Two drugs (proguanil and sulfasalazine) implicated in viral replication were shown to inhibit replication in cell assays. This unbiased and validated analysis opens new avenues for the rapid repurposing of approved drugs into clinical trials.


Subject(s)
Drug Repositioning , SARS-CoV-2/physiology , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19/pathology , COVID-19/virology , Humans , Neural Networks, Computer , Proguanil/pharmacology , Proguanil/therapeutic use , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Sulfasalazine/pharmacology , Virus Replication/drug effects , COVID-19 Drug Treatment
6.
Genes Dev ; 35(13-14): 1005-1019, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34168039

ABSTRACT

N6-methyladenosine (m6A) is an abundant internal RNA modification, influencing transcript fate and function in uninfected and virus-infected cells. Installation of m6A by the nuclear RNA methyltransferase METTL3 occurs cotranscriptionally; however, the genomes of some cytoplasmic RNA viruses are also m6A-modified. How the cellular m6A modification machinery impacts coronavirus replication, which occurs exclusively in the cytoplasm, is unknown. Here we show that replication of SARS-CoV-2, the agent responsible for the COVID-19 pandemic, and a seasonal human ß-coronavirus HCoV-OC43, can be suppressed by depletion of METTL3 or cytoplasmic m6A reader proteins YTHDF1 and YTHDF3 and by a highly specific small molecule METTL3 inhibitor. Reduction of infectious titer correlates with decreased synthesis of viral RNAs and the essential nucleocapsid (N) protein. Sites of m6A modification on genomic and subgenomic RNAs of both viruses were mapped by methylated RNA immunoprecipitation sequencing (meRIP-seq). Levels of host factors involved in m6A installation, removal, and recognition were unchanged by HCoV-OC43 infection; however, nuclear localization of METTL3 and cytoplasmic m6A readers YTHDF1 and YTHDF2 increased. This establishes that coronavirus RNAs are m6A-modified and host m6A pathway components control ß-coronavirus replication. Moreover, it illustrates the therapeutic potential of targeting the m6A pathway to restrict coronavirus reproduction.


Subject(s)
Coronavirus OC43, Human/physiology , RNA Processing, Post-Transcriptional/genetics , SARS-CoV-2/physiology , Virus Replication/genetics , Adenosine/analogs & derivatives , Adenosine/genetics , Adenosine/metabolism , Cell Line , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Gene Expression Regulation/drug effects , Host-Pathogen Interactions/drug effects , Humans , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Nucleocapsid Proteins , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Virus Replication/drug effects
7.
Mol Cell ; 81(13): 2793-2807.e8, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33979575

ABSTRACT

DNA replication initiates at genomic locations known as origins of replication, which, in S. cerevisiae, share a common DNA consensus motif. Despite being virtually nucleosome-free, origins of replication are greatly influenced by the surrounding chromatin state. Here, we show that histone H3 lysine 37 mono-methylation (H3K37me1) is catalyzed by Set1p and Set2p and that it regulates replication origin licensing. H3K37me1 is uniformly distributed throughout most of the genome, but it is scarce at replication origins, where it increases according to the timing of their firing. We find that H3K37me1 hinders Mcm2 interaction with chromatin, maintaining low levels of MCM outside of conventional replication origins. Lack of H3K37me1 results in defective DNA replication from canonical origins while promoting replication events at inefficient and non-canonical sites. Collectively, our results indicate that H3K37me1 ensures correct execution of the DNA replication program by protecting the genome from inappropriate origin licensing and spurious DNA replication.


Subject(s)
DNA Replication , DNA, Fungal/biosynthesis , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methyltransferases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DNA, Fungal/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Methylation , Methyltransferases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
8.
Nature ; 593(7860): 597-601, 2021 05.
Article in English | MEDLINE | ID: mdl-33902106

ABSTRACT

N6-methyladenosine (m6A) is an abundant internal RNA modification1,2 that is catalysed predominantly by the METTL3-METTL14 methyltransferase complex3,4. The m6A methyltransferase METTL3 has been linked to the initiation and maintenance of acute myeloid leukaemia (AML), but the potential of therapeutic applications targeting this enzyme remains unknown5-7. Here we present the identification and characterization of STM2457, a highly potent and selective first-in-class catalytic inhibitor of METTL3, and a crystal structure of STM2457 in complex with METTL3-METTL14. Treatment of tumours with STM2457 leads to reduced AML growth and an increase in differentiation and apoptosis. These cellular effects are accompanied by selective reduction of m6A levels on known leukaemogenic mRNAs and a decrease in their expression consistent with a translational defect. We demonstrate that pharmacological inhibition of METTL3 in vivo leads to impaired engraftment and prolonged survival in various mouse models of AML, specifically targeting key stem cell subpopulations of AML. Collectively, these results reveal the inhibition of METTL3 as a potential therapeutic strategy against AML, and provide proof of concept that the targeting of RNA-modifying enzymes represents a promising avenue for anticancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Methyltransferases/antagonists & inhibitors , Adenosine/analogs & derivatives , Animals , Apoptosis , Cell Differentiation , Cell Line, Tumor , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Xenograft Model Antitumor Assays
10.
Nat Rev Cancer ; 20(6): 303-322, 2020 06.
Article in English | MEDLINE | ID: mdl-32300195

ABSTRACT

Specific chemical modifications of biological molecules are an efficient way of regulating molecular function, and a plethora of downstream signalling pathways are influenced by the modification of DNA and proteins. Many of the enzymes responsible for regulating protein and DNA modifications are targets of current cancer therapies. RNA epitranscriptomics, the study of RNA modifications, is the new frontier of this arena. Despite being known since the 1970s, eukaryotic RNA modifications were mostly identified on transfer RNA and ribosomal RNA until the last decade, when they have been identified and characterized on mRNA and various non-coding RNAs. Increasing evidence suggests that RNA modification pathways are also misregulated in human cancers and may be ideal targets of cancer therapy. In this Review we highlight the RNA epitranscriptomic pathways implicated in cancer, describing their biological functions and their connections to the disease.


Subject(s)
Epigenesis, Genetic/genetics , Neoplasms/genetics , Neoplasms/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA/metabolism , Epigenesis, Genetic/physiology , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , RNA/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Nat Commun ; 11(1): 926, 2020 02 17.
Article in English | MEDLINE | ID: mdl-32066737

ABSTRACT

The field of epitranscriptomics continues to reveal how post-transcriptional modification of RNA affects a wide variety of biological phenomena. A pivotal challenge in this area is the identification of modified RNA residues within their sequence contexts. Mass spectrometry (MS) offers a comprehensive solution by using analogous approaches to shotgun proteomics. However, software support for the analysis of RNA MS data is inadequate at present and does not allow high-throughput processing. Existing software solutions lack the raw performance and statistical grounding to efficiently handle the numerous modifications found on RNA. We present a free and open-source database search engine for RNA MS data, called NucleicAcidSearchEngine (NASE), that addresses these shortcomings. We demonstrate the capability of NASE to reliably identify a wide range of modified RNA sequences in four original datasets of varying complexity. In human tRNA, we characterize over 20 different modification types simultaneously and find many cases of incomplete modification.


Subject(s)
Epigenomics/methods , High-Throughput Screening Assays/methods , RNA Processing, Post-Transcriptional/genetics , Search Engine , Tandem Mass Spectrometry/methods , Base Sequence/genetics , Databases, Factual/statistics & numerical data , Datasets as Topic , Humans , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Reproducibility of Results
12.
Nat Struct Mol Biol ; 26(10): 858-862, 2019 10.
Article in English | MEDLINE | ID: mdl-31582848

ABSTRACT

Exciting research has connected specific RNA modifications to chromatin, providing evidence for co-transcriptional deposition and function in gene regulation. Here we review insights gained from studying the co-transcriptional roles of RNA modifications, and their influence in normal and disease contexts. We also discuss how the availability of novel technical approaches could raise the translational potential of targeting RNA-modifying enzymes for the treatment of disease.


Subject(s)
Chromatin/metabolism , RNA Processing, Post-Transcriptional , RNA/metabolism , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly , Gene Expression Regulation , Humans , RNA/genetics , Transcription, Genetic
13.
Exp Cell Res ; 381(1): 129-138, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31077711

ABSTRACT

Sox2 is a master transcriptional regulator of embryonic development. In this study, we determined the protein interactome of Sox2 in the chromatin and nucleoplasm of mouse embryonic stem (mES) cells. Apart from canonical interactions with pluripotency-regulating transcription factors, we identified interactions with several chromatin modulators, including members of the heterochromatin protein 1 (HP1) family, suggesting a role for Sox2 in chromatin-mediated transcriptional repression. Sox2 was also found to interact with RNA binding proteins (RBPs), including proteins involved in RNA processing. RNA immunoprecipitation followed by sequencing revealed that Sox2 associates with different messenger RNAs, as well as small nucleolar RNA Snord34 and the non-coding RNA 7SK. 7SK has been shown to regulate transcription at gene regulatory regions, which could suggest a functional interaction with Sox2 for chromatin recruitment. Nevertheless, we found no evidence of Sox2 modulating recruitment of 7SK to chromatin when examining 7SK chromatin occupancy by Chromatin Isolation by RNA Purification (ChIRP) in Sox2 depleted mES cells. In addition, knockdown of 7SK in mES cells did not lead to any change in Sox2 occupancy at 7SK-regulated genes. Thus, our results show that Sox2 extensively interacts with RBPs, and suggest that Sox2 and 7SK co-exist in a ribonucleoprotein complex whose function is not to regulate chromatin recruitment, but could rather regulate other processes in the nucleoplasm.


Subject(s)
Mouse Embryonic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cell Line , Chromatin/metabolism , Gene Knockdown Techniques , Mice , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/genetics
14.
Epigenetics Chromatin ; 12(1): 21, 2019 Apr 02.
Article in English | MEDLINE | ID: mdl-30940194

ABSTRACT

BACKGROUND: Stem cell differentiation involves major chromatin reorganisation, heterochromatin formation and genomic relocalisation of structural proteins, including heterochromatin protein 1 gamma (HP1γ). As the principal reader of the repressive histone marks H3K9me2/3, HP1 plays a key role in numerous processes including heterochromatin formation and maintenance. RESULTS: We find that HP1γ is citrullinated in mouse embryonic stem cells (mESCs) and this diminishes when cells differentiate, indicating that it is a dynamically regulated post-translational modification during stem cell differentiation. Peptidylarginine deiminase 4, a known regulator of pluripotency, citrullinates HP1γ in vitro. This requires R38 and R39 within the HP1γ chromodomain, and the catalytic activity is enhanced by trimethylated H3K9 (H3K9me3) peptides. Mutation of R38 and R39, designed to mimic citrullination, affects HP1γ binding to H3K9me3-containing peptides. Using live-cell single-particle tracking, we demonstrate that R38 and R39 are important for HP1γ binding to chromatin in vivo. Furthermore, their mutation reduces the residence time of HP1γ on chromatin in differentiating mESCs. CONCLUSION: Citrullination is a novel post-translational modification of the structural heterochromatin protein HP1γ in mESCs that is dynamically regulated during mESC differentiation. The citrullinated residues lie within the HP1γ chromodomain and are important for H3K9me3 binding in vitro and chromatin association in vivo.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Citrullination , Heterochromatin/metabolism , Animals , Binding Sites , Cell Differentiation , Cell Line , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Heterochromatin/chemistry , Heterochromatin/genetics , Histone Code , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mutation , Protein Binding , Protein-Arginine Deiminases/metabolism
15.
Mol Cell ; 74(6): 1278-1290.e9, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31031083

ABSTRACT

7-methylguanosine (m7G) is present at mRNA caps and at defined internal positions within tRNAs and rRNAs. However, its detection within low-abundance mRNAs and microRNAs (miRNAs) has been hampered by a lack of sensitive detection strategies. Here, we adapt a chemical reactivity assay to detect internal m7G in miRNAs. Using this technique (Borohydride Reduction sequencing [BoRed-seq]) alongside RNA immunoprecipitation, we identify m7G within a subset of miRNAs that inhibit cell migration. We show that the METTL1 methyltransferase mediates m7G methylation within miRNAs and that this enzyme regulates cell migration via its catalytic activity. Using refined mass spectrometry methods, we map m7G to a single guanosine within the let-7e-5p miRNA. We show that METTL1-mediated methylation augments let-7 miRNA processing by disrupting an inhibitory secondary structure within the primary miRNA transcript (pri-miRNA). These results identify METTL1-dependent N7-methylation of guanosine as a new RNA modification pathway that regulates miRNA structure, biogenesis, and cell migration.


Subject(s)
Guanosine/analogs & derivatives , Methyltransferases/genetics , MicroRNAs/genetics , RNA Processing, Post-Transcriptional , A549 Cells , Base Sequence , Biological Assay , Caco-2 Cells , Cell Movement , Cell Proliferation , Guanosine/metabolism , HEK293 Cells , Humans , Methylation , Methyltransferases/metabolism , MicroRNAs/metabolism , Nucleic Acid Conformation
16.
Nat Commun ; 9(1): 5378, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30568163

ABSTRACT

We recently identified the splicing kinase gene SRPK1 as a genetic vulnerability of acute myeloid leukemia (AML). Here, we show that genetic or pharmacological inhibition of SRPK1 leads to cell cycle arrest, leukemic cell differentiation and prolonged survival of mice transplanted with MLL-rearranged AML. RNA-seq analysis demonstrates that SRPK1 inhibition leads to altered isoform levels of many genes including several with established roles in leukemogenesis such as MYB, BRD4 and MED24. We focus on BRD4 as its main isoforms have distinct molecular properties and find that SRPK1 inhibition produces a significant switch from the short to the long isoform at the mRNA and protein levels. This was associated with BRD4 eviction from genomic loci involved in leukemogenesis including BCL2 and MYC. We go on to show that this switch mediates at least part of the anti-leukemic effects of SRPK1 inhibition. Our findings reveal that SRPK1 represents a plausible new therapeutic target against AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Differentiation , Chromatin/metabolism , Epigenesis, Genetic , HL-60 Cells , Hematopoiesis , Humans , K562 Cells , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Splicing
17.
Mol Cell ; 72(4): 625-635.e4, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30454561

ABSTRACT

In response to genotoxic stress, cells activate a signaling cascade known as the DNA damage checkpoint (DDC) that leads to a temporary cell cycle arrest and activation of DNA repair mechanisms. Because persistent DDC activation compromises cell viability, this process must be tightly regulated. However, despite its importance, the mechanisms regulating DDC recovery are not completely understood. Here, we identify a DNA-damage-regulated histone modification in Saccharomyces cerevisiae, phosphorylation of H4 threonine 80 (H4T80ph), and show that it triggers checkpoint inactivation. H4T80ph is critical for cell survival to DNA damage, and its absence causes impaired DDC recovery and persistent cell cycle arrest. We show that, in response to genotoxic stress, p21-activated kinase Cla4 phosphorylates H4T80 to recruit Rtt107 to sites of DNA damage. Rtt107 displaces the checkpoint adaptor Rad9, thereby interrupting the checkpoint-signaling cascade. Collectively, our results indicate that H4T80ph regulates DDC recovery.


Subject(s)
DNA Damage , DNA Repair , Histones/genetics , Histones/metabolism , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
18.
Sci Signal ; 11(537)2018 07 03.
Article in English | MEDLINE | ID: mdl-29970603

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is an incurable premature aging disease. Identifying deregulated biological processes in HGPS might thus help define novel therapeutic strategies. Fibroblasts from HGPS patients display defects in nucleocytoplasmic shuttling of the GTP-bound form of the small GTPase Ran (RanGTP), which leads to abnormal transport of proteins into the nucleus. We report that microtubule stabilization in HGPS cells sequestered the nonclassical nuclear import protein Transportin-1 (TNPO1) in the cytoplasm, thus affecting the nuclear localization of its cargo, including the nuclear pore protein NUP153. Consequently, nuclear Ran, nuclear anchorage of the nucleoporin TPR, and chromatin organization were disrupted, deregulating gene expression and inducing senescence. Inhibiting N-acetyltransferase 10 (NAT10) ameliorated HGPS phenotypes by rebalancing the nuclear to cytoplasmic ratio of TNPO1. This restored nuclear pore complex integrity and nuclear Ran localization, thereby correcting HGPS cellular phenotypes. We observed a similar mechanism in cells from healthy aged individuals. This study identifies a nuclear import pathway affected in aging and underscores the potential for NAT10 inhibition as a possible therapeutic strategy for HGPS and perhaps also for pathologies associated with normal aging.


Subject(s)
Cell Nucleus/metabolism , Cellular Senescence , N-Terminal Acetyltransferase E/antagonists & inhibitors , Nuclear Pore Complex Proteins/metabolism , Progeria/prevention & control , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Adult , Aged, 80 and over , Case-Control Studies , Cells, Cultured , Child , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Microtubules/metabolism , Microtubules/pathology , N-Terminal Acetyltransferase E/genetics , N-Terminal Acetyltransferase E/metabolism , N-Terminal Acetyltransferases , Nuclear Pore Complex Proteins/genetics , Phenotype , Progeria/genetics , Progeria/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Young Adult , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
20.
FEBS Lett ; 592(13): 2308-2322, 2018 07.
Article in English | MEDLINE | ID: mdl-29782654

ABSTRACT

DDX3X is a multifunctional RNA helicase with documented roles in different cancer types. Here, we demonstrate that DDX3X plays an oncogenic role in breast cancer cells by modulating the cell cycle. Depletion of DDX3X in MCF7 cells slows cell proliferation by inducing a G1 phase arrest. Notably, DDX3X inhibits expression of Kruppel-like factor 4 (KLF4), a transcription factor and cell cycle repressor. Moreover, DDX3X directly interacts with KLF4 mRNA and regulates its splicing. We show that DDX3X-mediated repression of KLF4 promotes expression of S-phase inducing genes in MCF7 breast cancer cells. These findings provide evidence for a novel function of DDX3X in regulating expression and downstream functions of KLF4, a master negative regulator of the cell cycle.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle/genetics , Cell Proliferation/genetics , DEAD-box RNA Helicases/physiology , Kruppel-Like Transcription Factors/genetics , Breast Neoplasms/genetics , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4 , MCF-7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...