Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Cell ; 173(5): 1204-1216.e26, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29628141

ABSTRACT

Pseudouridylation (Ψ) is the most abundant and widespread type of RNA epigenetic modification in living organisms; however, the biological role of Ψ remains poorly understood. Here, we show that a Ψ-driven posttranscriptional program steers translation control to impact stem cell commitment during early embryogenesis. Mechanistically, the Ψ "writer" PUS7 modifies and activates a novel network of tRNA-derived small fragments (tRFs) targeting the translation initiation complex. PUS7 inactivation in embryonic stem cells impairs tRF-mediated translation regulation, leading to increased protein biosynthesis and defective germ layer specification. Remarkably, dysregulation of this posttranscriptional regulatory circuitry impairs hematopoietic stem cell commitment and is common to aggressive subtypes of human myelodysplastic syndromes. Our findings unveil a critical function of Ψ in directing translation control in stem cells with important implications for development and disease.


Subject(s)
Intramolecular Transferases/metabolism , Protein Biosynthesis , Pseudouridine/metabolism , RNA, Transfer/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Differentiation , Eukaryotic Initiation Factors/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Intramolecular Transferases/antagonists & inhibitors , Intramolecular Transferases/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Myelodysplastic Syndromes/pathology , Nucleic Acid Conformation , Phosphoproteins/metabolism , Poly(A)-Binding Protein I/antagonists & inhibitors , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Stem Cell Niche
2.
Cell ; 158(6): 1281-1292, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215487

ABSTRACT

A dichotomous choice for metazoan cells is between proliferation and differentiation. Measuring tRNA pools in various cell types, we found two distinct subsets, one that is induced in proliferating cells, and repressed otherwise, and another with the opposite signature. Correspondingly, we found that genes serving cell-autonomous functions and genes involved in multicellularity obey distinct codon usage. Proliferation-induced and differentiation-induced tRNAs often carry anticodons that correspond to the codons enriched among the cell-autonomous and the multicellularity genes, respectively. Because mRNAs of cell-autonomous genes are induced in proliferation and cancer in particular, the concomitant induction of their codon-enriched tRNAs suggests coordination between transcription and translation. Histone modifications indeed change similarly in the vicinity of cell-autonomous genes and their corresponding tRNAs, and in multicellularity genes and their tRNAs, suggesting the existence of transcriptional programs coordinating tRNA supply and demand. Hence, we describe the existence of two distinct translation programs that operate during proliferation and differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Protein Biosynthesis , RNA, Transfer/genetics , Anticodon , Cell Line, Tumor , Cell Transformation, Neoplastic , Codon , Histones/metabolism , Humans , Neoplasms/genetics , RNA, Messenger/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Transcriptome
3.
EMBO J ; 42(4): e112835, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36695748

ABSTRACT

In this commentary, Sonne-Hansen and colleagues argue that research leaders and organizations should encourage more "theory-guessing" by budding young scientists, rather than incentivizing safe mainstream research.


Subject(s)
Antidotes , Creativity
4.
Trends Biochem Sci ; 47(1): 66-81, 2022 01.
Article in English | MEDLINE | ID: mdl-34312084

ABSTRACT

The conceptual origins of ribosome specialization can be traced back to the earliest days of molecular biology. Yet, this field has only recently begun to gather momentum, with numerous studies identifying distinct heterogeneous ribosome populations across multiple species and model systems. It is proposed that some of these compositionally distinct ribosomes may be functionally specialized and able to regulate the translation of specific mRNAs. Identification and functional characterization of specialized ribosomes has the potential to elucidate a novel layer of gene expression control, at the level of translation, where the ribosome itself is a key regulatory player. In this review, we discuss different sources of ribosome heterogeneity, evidence for ribosome specialization, and also the future directions of this exciting field.


Subject(s)
Protein Biosynthesis , Ribosomal Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/metabolism , Ribosomal Proteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism
5.
Blood ; 139(2): 245-255, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34359076

ABSTRACT

Novel therapies for the treatment of acute myeloid leukemia (AML) are urgently needed, because current treatments do not cure most patients with AML. We report a domain-focused, kinome-wide CRISPR-Cas9 screening that identified protein kinase targets for the treatment of AML, which led to the identification of Rio-kinase 2 (RIOK2) as a potential novel target. Loss of RIOK2 led to a decrease in protein synthesis and to ribosomal instability followed by apoptosis in leukemic cells, but not in fibroblasts. Moreover, the ATPase function of RIOK2 was necessary for cell survival. When a small-molecule inhibitor was used, pharmacological inhibition of RIOK2 similarly led to loss of protein synthesis and apoptosis and affected leukemic cell growth in vivo. Our results provide proof of concept for targeting RIOK2 as a potential treatment of patients with AML.


Subject(s)
Leukemia, Myeloid, Acute , Protein Kinase Inhibitors , Animals , Mice , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , CRISPR-Cas Systems , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Molecular Targeted Therapy , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology
6.
RNA ; 26(9): 1104-1117, 2020 09.
Article in English | MEDLINE | ID: mdl-32393525

ABSTRACT

Noncoding RNA has a proven ability to direct and regulate chromatin modifications by acting as scaffolds between DNA and histone-modifying complexes. However, it is unknown if ncRNA plays any role in DNA replication and epigenome maintenance, including histone eviction and reinstallment of histone modifications after genome duplication. Isolation of nascent chromatin has identified a large number of RNA-binding proteins in addition to unknown components of the replication and epigenetic maintenance machinery. Here, we isolated and characterized long and short RNAs associated with nascent chromatin at active replication forks and track RNA composition during chromatin maturation across the cell cycle. Shortly after fork passage, GA-rich-, alpha- and TElomeric Repeat-containing RNAs (TERRA) are associated with replicated DNA. These repeat containing RNAs arise from loci undergoing replication, suggesting an interaction in cis. Post-replication during chromatin maturation, and even after mitosis in G1, the repeats remain enriched on DNA. This suggests that specific types of repeat RNAs are transcribed shortly after DNA replication and stably associate with their loci of origin throughout the cell cycle. The presented method and data enable studies of RNA interactions with replication forks and post-replicative chromatin and provide insights into how repeat RNAs and their engagement with chromatin are regulated with respect to DNA replication and across the cell cycle.


Subject(s)
DNA Replication/genetics , DNA/genetics , Protein Processing, Post-Translational/genetics , RNA/genetics , Cell Cycle/genetics , Cell Line, Tumor , Chromatin/genetics , HeLa Cells , Histones/genetics , Humans
7.
EMBO Rep ; 19(6)2018 06.
Article in English | MEDLINE | ID: mdl-29712776

ABSTRACT

Autophagy is an essential catabolic process responsible for recycling of intracellular material and preserving cellular fidelity. Key to the autophagy pathway is the ubiquitin-like conjugation system mediating lipidation of Atg8 proteins and their anchoring to autophagosomal membranes. While regulation of autophagy has been characterized at the level of transcription, protein interactions and post-translational modifications, its translational regulation remains elusive. Here we describe a role for the conserved eukaryotic translation initiation factor 5A (eIF5A) in autophagy. Identified from a high-throughput screen, we find that eIF5A is required for lipidation of LC3B and its paralogs and promotes autophagosome formation. This feature is evolutionarily conserved and results from the translation of the E2-like ATG3 protein. Mechanistically, we identify an amino acid motif in ATG3 causing eIF5A dependency for its efficient translation. Our study identifies eIF5A as a key requirement for autophagosome formation and demonstrates the importance of translation in mediating efficient autophagy.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Peptide Initiation Factors/physiology , Protein Biosynthesis , RNA-Binding Proteins/physiology , Ubiquitin-Conjugating Enzymes/metabolism , Autophagy-Related Proteins/genetics , Humans , MCF-7 Cells , Microtubule-Associated Proteins/metabolism , Protein Processing, Post-Translational , Proteomics , Ubiquitin-Conjugating Enzymes/genetics , Eukaryotic Translation Initiation Factor 5A
8.
Nucleic Acids Res ; 46(1): e4, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29059327

ABSTRACT

Deciphering the functions of long non-coding RNAs (lncRNAs) is facilitated by visualization of their subcellular localization using in situ hybridization (ISH) techniques. We evaluated four different ISH methods for detection of MALAT1 and CYTOR in cultured cells: a multiple probe detection approach with or without enzymatic signal amplification, a branched-DNA (bDNA) probe and an LNA-modified probe with enzymatic signal amplification. All four methods adequately stained MALAT1 in the nucleus in all of three cell lines investigated, HeLa, NHDF and T47D, and three of the methods detected the less expressed CYTOR. The sensitivity of the four ISH methods was evaluated by image analysis. In all three cell lines, the two methods involving enzymatic amplification gave the most intense MALAT1 signal, but the signal-to-background ratios were not different. CYTOR was best detected using the bDNA method. All four ISH methods showed significantly reduced MALAT1 signal in knock-out cells, and siRNA-induced knock-down of CYTOR resulted in significantly reduced CYTOR ISH signal, indicating good specificity of the probe designs and detection systems. Our data suggest that the ISH methods allow detection of both abundant and less abundantly expressed lncRNAs, although the latter required the use of the most specific and sensitive probe detection system.


Subject(s)
Cell Nucleus/genetics , Gene Expression Regulation , In Situ Hybridization, Fluorescence/methods , RNA, Long Noncoding/genetics , A549 Cells , Cell Line , Cell Line, Tumor , DNA Probes/genetics , Gene Amplification , HeLa Cells , Humans , MCF-7 Cells , Reproducibility of Results
9.
Semin Cell Dev Biol ; 65: 1-10, 2017 05.
Article in English | MEDLINE | ID: mdl-27438587

ABSTRACT

The striking similarities between pluripotent and cancer cells, such as immortality and increased stress resistance, have long been acknowledged. Numerous studies searched for and successfully identified common molecular players and pathways, thus providing an entirely new challenge and potential therapeutic angle by targeting cancer cells or a specific stem population of the tumor via pluripotency associated processes. However, these strategies have until now mainly been restricted to proteins. Nonetheless, it has become clear over the past decade that the overwhelming majority of the genome produces noncoding transcripts, many of which have proven both functional and crucial for key cellular processes, including stemness maintenance. Moreover, numerous long noncoding RNAs are deregulated in cancer, but little is known concerning their functions and molecular mechanisms. Consequently, it seems essential to integrate the noncoding transcripts into the picture of the stemness-cancer connection. Whereas a number of studies have addressed the expression of lncRNAs in cancer stem cells, no systematic approach has yet been undertaken to identify lncRNAs implicated in the maintenance of the embryonic stemness state that is hijacked by cancer cells. The aim of this review is to highlight long noncoding RNAs with shared functions in stemness and cancer and to outline the current state of a field in its infancy, the search for long noncoding transcripts in cancer stem cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , RNA, Long Noncoding/genetics , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Genetic Vectors/therapeutic use , Humans , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/pathology , RNA, Long Noncoding/agonists , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , Signal Transduction
10.
Anal Chem ; 90(24): 14198-14206, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30422637

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell non-Hodgkin lymphoma. To treat this aggressive disease, R-CHOP, a combination of immunotherapy (R; rituximab) and chemotherapy (CHOP; cyclophosphamide, doxorubicin, vincristine, and prednisone), remains the most commonly used regimen for newly diagnosed DLBCLs. However, up to one-third of patients ultimately becomes refractory to initial therapy or relapses after treatment, and the high mortality rate highlights the urgent need for novel therapeutic approaches based upon selective molecular targets. In order to understand the molecular mechanisms underlying relapsed DLBCL, we studied differences in the lipid and metabolic composition of nontreated and R-CHOP-resistant tumors, using a combination of in vivo DLBCL xenograft models and mass spectrometry imaging. Together, these techniques provide information regarding analyte composition and molecular distributions of therapy-resistant and sensitive areas. We found specific lipid and metabolic profiles for R-CHOP-resistant tumors, such as a higher presence of phosphatidylinositol and sphingomyelin fragments. In addition, we investigated intratumor heterogeneity and identified specific lipid markers of viable and necrotic areas. Furthermore, we could monitor metabolic changes and found reduced adenosine triphosphate and increased adenosine monophosphate in the R-CHOP-resistant tumors. This work highlights the power of combining in vivo imaging and MSI to track molecular signatures in DLBCL, which has potential application for other diseases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lipids/analysis , Lymphoma, Large B-Cell, Diffuse/drug therapy , Metabolome , Rituximab/therapeutic use , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Discriminant Analysis , Drug Resistance, Neoplasm , Humans , Luminescent Measurements , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Neoplasm Recurrence, Local , Phosphatidylinositols/analysis , Principal Component Analysis , Transplantation, Heterologous
11.
Nucleic Acids Res ; 44(16): 7884-95, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27257078

ABSTRACT

Ribose methylation is one of the two most abundant modifications in human ribosomal RNA and is believed to be important for ribosome biogenesis, mRNA selectivity and translational fidelity. We have applied RiboMeth-seq to rRNA from HeLa cells for ribosome-wide, quantitative mapping of 2'-O-Me sites and obtained a comprehensive set of 106 sites, including two novel sites, and with plausible box C/D guide RNAs assigned to all but three sites. We find approximately two-thirds of the sites to be fully methylated and the remainder to be fractionally modified in support of ribosome heterogeneity at the level of RNA modifications. A comparison to HCT116 cells reveals similar 2'-O-Me profiles with distinct differences at several sites. This study constitutes the first comprehensive mapping of 2'-O-Me sites in human rRNA using a high throughput sequencing approach. It establishes the existence of a core of constitutively methylated positions and a subset of variable, potentially regulatory positions, and paves the way for experimental analyses of the role of variations in rRNA methylation under different physiological or pathological settings.


Subject(s)
RNA, Ribosomal/metabolism , Ribosomes/metabolism , Base Sequence , HCT116 Cells , HeLa Cells , Humans , Methylation , RNA, Small Nucleolar/metabolism , Reproducibility of Results , Sequence Analysis, RNA
12.
Hum Mol Genet ; 24(23): 6565-79, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26395458

ABSTRACT

Type 2 brittle cornea syndrome (BCS2) is an inherited connective tissue disease with a devastating ocular phenotype caused by mutations in the transcription factor PR domain containing 5 (PRDM5) hypothesized to exert epigenetic effects through histone and DNA methylation. Here we investigate clinical samples, including skin fibroblasts and retinal tissue from BCS2 patients, to elucidate the epigenetic role of PRDM5 and mechanisms of its dysregulation in disease. First we report abnormal retinal vascular morphology in the eyes of two cousins with BCS2 (PRDM5 Δ exons 9-14) using immunohistochemistry, and mine data from skin fibroblast expression microarrays from patients with PRDM5 mutations p.Arg590* and Δ exons 9-14, as well as from a PRDM5 ChIP-sequencing experiment. Gene ontology analysis of dysregulated PRDM5-target genes reveals enrichment for extracellular matrix (ECM) genes supporting vascular integrity and development. Q-PCR and ChIP-qPCR confirm upregulation of critical mediators of ECM stability in vascular structures (COL13A1, COL15A1, NTN1, CDH5) in patient fibroblasts. We identify H3K9 di-methylation (H3K9me2) at these PRDM5-target genes in fibroblasts, and demonstrate that the BCS2 mutation p.Arg83Cys diminishes interaction of PRDM5 with repressive complexes, including NuRD complex protein CHD4, and the repressive chromatin interactor HP1BP3, by co-immunoprecipitation combined with mass spectrometry. We observe reduced heterochromatin protein 1 binding protein 3 (HP1BP3) staining in the retinas of two cousins lacking exons 9-14 by immunohistochemistry, and dysregulated H3K9me2 in skin fibroblasts of three patients (p.Arg590*, p.Glu134* and Δ exons 9-14) by western blotting. These findings suggest that defective interaction of PRDM5 with repressive complexes, and dysregulation of H3K9me2, play a role in PRDM5-associated disease.


Subject(s)
DNA-Binding Proteins/genetics , Ehlers-Danlos Syndrome/genetics , Histones/metabolism , Mutation , Retinal Vessels/pathology , Transcription Factors/genetics , Adult , Antigens, CD/genetics , Cadherins/genetics , Child , Collagen/genetics , Ehlers-Danlos Syndrome/metabolism , Ehlers-Danlos Syndrome/pathology , Female , Fibroblasts/metabolism , Gene Ontology , Humans , Male , Methylation , Middle Aged , Nerve Growth Factors/genetics , Netrin-1 , Skin/cytology , Tumor Suppressor Proteins/genetics , Up-Regulation , Young Adult
13.
Nucleic Acids Res ; 43(13): 6207-21, 2015 07 27.
Article in English | MEDLINE | ID: mdl-26089393

ABSTRACT

We report a high-resolution time series study of transcriptome dynamics following antimiR-mediated inhibition of miR-9 in a Hodgkin lymphoma cell-line-the first such dynamic study of the microRNA inhibition response-revealing both general and specific aspects of the physiological response. We show miR-9 inhibition inducing a multiphasic transcriptome response, with a direct target perturbation before 4 h, earlier than previously reported, amplified by a downstream peak at ∼32 h consistent with an indirect response due to secondary coherent regulation. Predictive modelling indicates a major role for miR-9 in post-transcriptional control of RNA processing and RNA binding protein regulation. Cluster analysis identifies multiple co-regulated gene regulatory modules. Functionally, we observe a shift over time from mRNA processing at early time points to translation at later time points. We validate the key observations with independent time series qPCR and we experimentally validate key predicted miR-9 targets. Methodologically, we developed sensitive functional data analytic predictive methods to analyse the weak response inherent in microRNA inhibition experiments. The methods of this study will be applicable to similar high-resolution time series transcriptome analyses and provides the context for more accurate experimental design and interpretation of future microRNA inhibition studies.


Subject(s)
Gene Expression Regulation , MicroRNAs/antagonists & inhibitors , Transcriptome , Cell Line, Tumor , Cluster Analysis , Genomics , Humans , Models, Genetic , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism
14.
RNA ; 20(2): 236-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344320

ABSTRACT

The past decade has shown mammalian genomes to be pervasively transcribed and identified thousands of noncoding (nc) transcripts. It is currently unclear to what extent these transcripts are of functional importance, as experimental functional evidence exists for only a small fraction. Here, we characterize the expression and evolutionary conservation properties of 12,115 known and novel nc transcripts, including structural RNAs, long nc RNAs (lncRNAs), antisense RNAs, EvoFold predictions, ultraconserved elements, and expressed nc regions. Expression levels are evaluated across 12 human tissues using a custom-designed microarray, supplemented with RNAseq. Conservation levels are evaluated at both the base level and at the syntenic level. We combine these measures with epigenetic mark annotations to identify subsets of novel nc transcripts that show characteristics similar to known functional ncRNAs. Few novel nc transcripts show both high expression and conservation levels. However, overall, we observe a positive correlation between expression and both conservation and epigenetic annotations, suggesting that a subset of the expressed transcripts are under purifying selection and likely functional. The identified subsets of expressed and conserved novel nc transcripts may form the basis for further functional characterization.


Subject(s)
RNA, Untranslated/genetics , Transcriptome , Base Sequence , Chromatin/genetics , Conserved Sequence , Expressed Sequence Tags , Humans , Inverted Repeat Sequences , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Open Reading Frames , Organ Specificity , RNA, Untranslated/metabolism
15.
Mol Cell ; 30(4): 460-71, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18498749

ABSTRACT

MicroRNAs (miRNAs) are small RNAs that function as posttranscriptional regulators of gene expression. miRNAs affect a variety of signaling pathways, and impaired miRNA regulation may contribute to the development of cancer and other diseases. Here we show that miRNA miR-10a interacts with the 5' untranslated region of mRNAs encoding ribosomal proteins to enhance their translation. miR-10a alleviates translational repression of the ribosomal protein mRNAs during amino acid starvation and is required for their translational induction following anisomycin treatment or overexpression of RAS. We show that miR-10a binds immediately downstream of the regulatory 5'TOP motif and that the 5'TOP regulatory complex and miR-10a are functionally interconnected. The results show that miR-10a may positively control global protein synthesis via the stimulation of ribosomal protein mRNA translation and ribosome biogenesis and hereby affect the ability of cells to undergo transformation.


Subject(s)
5' Untranslated Regions , Membrane Glycoproteins/metabolism , Protein Biosynthesis , Receptors, Immunologic/metabolism , Ribosomal Proteins/genetics , Amino Acids/metabolism , Animals , Base Sequence , Binding Sites , Cell Transformation, Neoplastic , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Mice , Molecular Sequence Data , NIH 3T3 Cells , Rats , Receptors, Immunologic/genetics , Ribosomal Proteins/metabolism
16.
PLoS Genet ; 9(10): e1003913, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204315

ABSTRACT

miRNAs are small regulatory RNAs that, due to their considerable potential to target a wide range of mRNAs, are implicated in essentially all biological process, including cancer. miR-10a is particularly interesting considering its conserved location in the Hox cluster of developmental regulators. A role for this microRNA has been described in developmental regulation as well as for various cancers. However, previous miR-10a studies are exclusively based on transient knockdowns of this miRNA and to extensively study miR-10a loss we have generated a miR-10a knock out mouse. Here we show that, in the Apc(min) mouse model of intestinal neoplasia, female miR-10a deficient mice develop significantly more adenomas than miR-10(+/+) and male controls. We further found that Lpo is extensively upregulated in the intestinal epithelium of mice deprived of miR-10a. Using in vitro assays, we demonstrate that the primary miR-10a target KLF4 can upregulate transcription of Lpo, whereas siRNA knockdown of KLF4 reduces LPO levels in HCT-116 cells. Furthermore, Klf4 is upregulated in the intestines of miR-10a knockout mice. Lpo has previously been shown to have the capacity to oxidize estrogens into potent depurinating mutagens, creating an instable genomic environment that can cause initiation of cancer. Therefore, we postulate that Lpo upregulation in the intestinal epithelium of miR-10a deficient mice together with the predominant abundance of estrogens in female animals mainly accounts for the sex-related cancer phenotype we observed. This suggests that miR-10a could be used as a potent diagnostic marker for discovering groups of women that are at high risk of developing colorectal carcinoma, which today is one of the leading causes of cancer-related deaths.


Subject(s)
Intestinal Neoplasms/genetics , Kruppel-Like Transcription Factors/biosynthesis , Lactoperoxidase/genetics , MicroRNAs/genetics , Animals , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Intestinal Neoplasms/pathology , Kruppel-Like Factor 4 , Lactoperoxidase/biosynthesis , Male , Mice , Mice, Knockout , MicroRNAs/metabolism , Wnt Signaling Pathway/genetics
17.
EMBO J ; 30(22): 4628-41, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21915098

ABSTRACT

Autophagy is an evolutionarily conserved mechanism of cellular self-digestion in which proteins and organelles are degraded through delivery to lysosomes. Defects in this process are implicated in numerous human diseases including cancer. To further elucidate regulatory mechanisms of autophagy, we performed a functional screen in search of microRNAs (miRNAs), which regulate the autophagic flux in breast cancer cells. In this study, we identified the tumour suppressive miRNA, miR-101, as a potent inhibitor of basal, etoposide- and rapamycin-induced autophagy. Through transcriptome profiling, we identified three novel miR-101 targets, STMN1, RAB5A and ATG4D. siRNA-mediated depletion of these genes phenocopied the effect of miR-101 overexpression, demonstrating their importance in autophagy regulation. Importantly, overexpression of STMN1 could partially rescue cells from miR-101-mediated inhibition of autophagy, indicating a functional importance for this target. Finally, we show that miR-101-mediated inhibition of autophagy can sensitize breast cancer cells to 4-hydroxytamoxifen (4-OHT)-mediated cell death. Collectively, these data establish a novel link between two highly important and rapidly growing research fields and present a new role for miR-101 as a key regulator of autophagy.


Subject(s)
Autophagy , MicroRNAs/genetics , MicroRNAs/metabolism , Stathmin/metabolism , Autophagy-Related Proteins , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Etoposide/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Small Interfering , Sirolimus/pharmacology , Stathmin/biosynthesis , Stathmin/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
18.
Bioessays ; 34(1): 50-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22028065

ABSTRACT

The PRDM family has recently spawned considerable interest as it has been implicated in fundamental aspects of cellular differentiation and exhibits expanding ties to human diseases. The PRDMs belong to the SET domain family of histone methyltransferases, however, enzymatic activity has been determined for only few PRDMs suggesting that they act by recruiting co-factors or, more speculatively, confer methylation of non-histone targets. Several PRDM family members are deregulated in human diseases, most prominently in hematological malignancies and solid cancers, where they can act as both tumor suppressors or drivers of oncogenic processes. The molecular mechanisms have been delineated for only few PRDMs and little is known about functional redundancy within the family. Future studies should identify target genes of PRDM proteins and the protein complexes in which PRDM proteins reside to provide a more comprehensive understanding of the biological and biochemical functions of this important protein family.


Subject(s)
Gene Expression Regulation , Hematologic Neoplasms/enzymology , Isoenzymes/metabolism , Neoplasms/enzymology , Protein Methyltransferases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Adipose Tissue, Brown/metabolism , Animals , Blood Vessels/metabolism , Cell Differentiation , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Hematopoiesis/physiology , Histones/genetics , Histones/metabolism , Humans , Isoenzymes/genetics , Methylation , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/pathology , Organ Specificity , Protein Methyltransferases/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics
19.
Cell Rep ; 43(4): 113998, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38551960

ABSTRACT

RNase L is an endoribonuclease of higher vertebrates that functions in antiviral innate immunity. Interferons induce oligoadenylate synthetase enzymes that sense double-stranded RNA of viral origin leading to the synthesis of 2',5'-oligoadenylate (2-5A) activators of RNase L. However, it is unknown precisely how RNase L remodels the host cell transcriptome. To isolate effects of RNase L from other effects of double-stranded RNA or virus, 2-5A is directly introduced into cells. Here, we report that RNase L activation by 2-5A causes a ribotoxic stress response involving the MAP kinase kinase kinase (MAP3K) ZAKα, MAP2Ks, and the stress-activated protein kinases JNK and p38α. RNase L activation profoundly alters the transcriptome by widespread depletion of mRNAs associated with different cellular functions but also by JNK/p38α-stimulated induction of inflammatory genes. These results show that the 2-5A/RNase L system triggers a protein kinase cascade leading to proinflammatory signaling and apoptosis.


Subject(s)
Endoribonucleases , Immunity, Innate , Endoribonucleases/metabolism , Endoribonucleases/genetics , Humans , Adenine Nucleotides/metabolism , Oligoribonucleotides/metabolism , Animals , Stress, Physiological , Transcriptome/genetics , RNA, Double-Stranded/metabolism
20.
FEBS Lett ; 598(7): 719-724, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38514456

ABSTRACT

The diverse range of organizations contributing to the global research ecosystem is believed to enhance the overall quality and resilience of its output. Mid-sized autonomous research institutes, distinct from universities, play a crucial role in this landscape. They often lead the way in new research fields and experimental methods, including those in social and organizational domains, which are vital for driving innovation. The EU-LIFE alliance was established with the goal of fostering excellence by developing and disseminating best practices among European biomedical research institutes. As directors of the 15 EU-LIFE institutes, we have spent a decade comparing and refining our processes. Now, we are eager to share the insights we've gained. To this end, we have crafted this Charter, outlining 10 principles we deem essential for research institutes to flourish and achieve ground-breaking discoveries. These principles, detailed in the Charter, encompass excellence, independence, training, internationality and inclusivity, mission focus, technological advancement, administrative innovation, cooperation, societal impact, and public engagement. Our aim is to inspire the establishment of new institutes that adhere to these principles and to raise awareness about their significance. We are convinced that they should be viewed a crucial component of any national and international innovation strategies.


Subject(s)
Biological Science Disciplines , Biomedical Research , Academies and Institutes
SELECTION OF CITATIONS
SEARCH DETAIL