Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Cell ; 186(19): 4100-4116.e15, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37643610

ABSTRACT

Nucleosomes block access to DNA methyltransferase, unless they are remodeled by DECREASE in DNA METHYLATION 1 (DDM1LSH/HELLS), a Snf2-like master regulator of epigenetic inheritance. We show that DDM1 promotes replacement of histone variant H3.3 by H3.1. In ddm1 mutants, DNA methylation is partly restored by loss of the H3.3 chaperone HIRA, while the H3.1 chaperone CAF-1 becomes essential. The single-particle cryo-EM structure at 3.2 Å of DDM1 with a variant nucleosome reveals engagement with histone H3.3 near residues required for assembly and with the unmodified H4 tail. An N-terminal autoinhibitory domain inhibits activity, while a disulfide bond in the helicase domain supports activity. DDM1 co-localizes with H3.1 and H3.3 during the cell cycle, and with the DNA methyltransferase MET1Dnmt1, but is blocked by H4K16 acetylation. The male germline H3.3 variant MGH3/HTR10 is resistant to remodeling by DDM1 and acts as a placeholder nucleosome in sperm cells for epigenetic inheritance.


Subject(s)
Arabidopsis Proteins , Arabidopsis , DNA Methylation , Histones , Nucleosomes , Chromatin Assembly and Disassembly , DNA , DNA Modification Methylases , Epigenesis, Genetic , Histones/genetics , Nucleosomes/genetics , Semen , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism
2.
Mol Cell ; 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39368465

ABSTRACT

MicroRNA (miRNA) biogenesis is initiated upon cleavage of a primary miRNA (pri-miRNA) hairpin by the Microprocessor (MP), composed of the Drosha RNase III enzyme and its partner DGCR8. Multiple pri-miRNA sequence motifs affect MP recognition, fidelity, and efficiency. Here, we performed cryoelectron microscopy (cryo-EM) and biochemical studies of several let-7 family pri-miRNAs in complex with human MP. We show that MP has the structural plasticity to accommodate a range of pri-miRNAs. These structures revealed key features of the 5' UG sequence motif, more comprehensively represented as the "flipped U with paired N" (fUN) motif. Our analysis explains how cleavage of class-II pri-let-7 members harboring a bulged nucleotide generates a non-canonical precursor with a 1-nt 3' overhang. Finally, the MP-SRSF3-pri-let-7f1 structure reveals how SRSF3 contributes to MP fidelity by interacting with the CNNC motif and Drosha's Piwi/Argonaute/Zwille (PAZ)-like domain. Overall, this study sheds light on the mechanisms for flexible recognition, accurate cleavage, and regulated processing of different pri-miRNAs by MP.

3.
Genes Dev ; 37(15-16): 743-759, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37669874

ABSTRACT

Protein tyrosine phosphatases (PTPs) are critical regulators of signal transduction but have yet to be exploited fully for drug development. Receptor protein tyrosine phosphatase δ (RPTPδ/PTPRD) has been shown to elicit tumor-promoting functions, including elevating SRC activity and promoting metastasis in certain cell contexts. Dimerization has been implicated in the inhibition of receptor protein tyrosine phosphatases (RPTPs). We have generated antibodies targeting PTPRD ectodomains with the goal of manipulating their dimerization status ectopically, thereby regulating intracellular signaling. We have validated antibody binding to endogenous PTPRD in a metastatic breast cancer cell line, CAL51, and demonstrated that a monoclonal antibody, RD-43, inhibited phosphatase activity and induced the degradation of PTPRD. Similar effects were observed following chemically induced dimerization of its phosphatase domain. Mechanistically, RD-43 triggered the formation of PTPRD dimers in which the phosphatase activity was impaired. Subsequently, the mAb-PTPRD dimer complex was degraded through lysosomal and proteasomal pathways, independently of secretase cleavage. Consequently, treatment with RD-43 inhibited SRC signaling and suppressed PTPRD-dependent cell invasion. Together, these findings demonstrate that manipulating RPTP function via antibodies to the extracellular segments has therapeutic potential.


Subject(s)
Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Signal Transduction , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Dimerization , Cell Line , Phosphoric Monoester Hydrolases
4.
Cell ; 160(4): 644-658, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25640237

ABSTRACT

Transcription in eukaryotes produces a number of long noncoding RNAs (lncRNAs). Two of these, MALAT1 and Menß, generate a tRNA-like small RNA in addition to the mature lncRNA. The stability of these tRNA-like small RNAs and bona fide tRNAs is monitored by the CCA-adding enzyme. Whereas CCA is added to stable tRNAs and tRNA-like transcripts, a second CCA repeat is added to certain unstable transcripts to initiate their degradation. Here, we characterize how these two scenarios are distinguished. Following the first CCA addition cycle, nucleotide binding to the active site triggers a clockwise screw motion, producing torque on the RNA. This ejects stable RNAs, whereas unstable RNAs are refolded while bound to the enzyme and subjected to a second CCA catalytic cycle. Intriguingly, with the CCA-adding enzyme acting as a molecular vise, the RNAs proofread themselves through differential responses to its interrogation between stable and unstable substrates.


Subject(s)
Archaeoglobus fulgidus/enzymology , Mitochondria/enzymology , RNA Nucleotidyltransferases/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Archaeoglobus fulgidus/metabolism , Base Sequence , Catalytic Domain , Humans , Mitochondria/metabolism , Models, Molecular , Nucleic Acid Conformation , RNA Nucleotidyltransferases/chemistry , RNA Nucleotidyltransferases/genetics , RNA Stability , RNA, Small Untranslated/metabolism
5.
Nature ; 607(7917): 169-175, 2022 07.
Article in English | MEDLINE | ID: mdl-35576971

ABSTRACT

Tuft cells are a rare chemosensory lineage that coordinates immune and neural responses to foreign pathogens in mucosal tissues1. Recent studies have also revealed tuft-cell-like human tumours2,3, particularly as a variant of small-cell lung cancer. Both normal and neoplastic tuft cells share a genetic requirement for the transcription factor POU2F3 (refs. 2,4), although the transcriptional mechanisms that generate this cell type are poorly understood. Here we show that binding of POU2F3 to the uncharacterized proteins C11orf53 and COLCA2 (renamed here OCA-T1/POU2AF2 and OCA-T2/POU2AF3, respectively) is critical in the tuft cell lineage. OCA-T1 and OCA-T2 are paralogues of the B-cell-specific coactivator OCA-B; all three proteins are encoded in a gene cluster and contain a conserved peptide that binds to class II POU transcription factors and a DNA octamer motif in a bivalent manner. We demonstrate that binding between POU2F3 and OCA-T1 or OCA-T2 is essential in tuft-cell-like small-cell lung cancer. Moreover, we generated OCA-T1-deficient mice, which are viable but lack tuft cells in several mucosal tissues. These findings reveal that the POU2F3-OCA-T complex is the master regulator of tuft cell identity and a molecular vulnerability of tuft-cell-like small-cell lung cancer.


Subject(s)
Cell Lineage , Lung Neoplasms , Neoplasm Proteins , Octamer Transcription Factors , Small Cell Lung Carcinoma , Animals , Humans , Mice , Lung Neoplasms/pathology , Mucous Membrane/pathology , Multigene Family/genetics , Neoplasm Proteins/metabolism , Nucleotide Motifs , Octamer Transcription Factors/metabolism , POU Domain Factors/metabolism , Small Cell Lung Carcinoma/pathology , Trans-Activators
6.
Cell ; 150(1): 100-10, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22682761

ABSTRACT

Argonaute proteins lie at the heart of the RNA-induced silencing complex (RISC), wherein they use small RNA guides to recognize targets. Initial insight into the architecture of Argonautes came from studies of prokaryotic proteins, revealing a crescent-shaped base made up of the amino-terminal, PAZ, middle, and PIWI domains. The recently reported crystal structure of human Argonaute-2 (hAgo2), the "slicer" in RNA interference, in complex with a mixed population of RNAs derived from insect cells provides insight into the architecture of a eukaryotic Argonaute protein with defined biochemical and biological functions. Here, we report the structure of human Ago2 bound to a physiologically relevant microRNA, microRNA-20a, at 2.2 Å resolution. The miRNA is anchored at both ends by the Mid and PAZ domains and makes several kinks and turns along the binding groove. Interestingly, miRNA binding confers remarkable stability on hAgo2, locking this otherwise flexible enzyme into a stable conformation.


Subject(s)
Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , MicroRNAs/chemistry , MicroRNAs/metabolism , Argonaute Proteins/isolation & purification , Crystallography, X-Ray , Humans , Models, Molecular , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
7.
Mol Cell ; 73(4): 845-856.e5, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30712989

ABSTRACT

ADP-ribosylation refers to the addition of one or more ADP-ribose groups onto proteins. The attached ADP-ribose monomers or polymers, commonly known as poly(ADP-ribose) (PAR), modulate the activities of the modified substrates or their binding affinities to other proteins. However, progress in this area is hindered by a lack of tools to investigate this protein modification. Here, we describe a new method named ELTA (enzymatic labeling of terminal ADP-ribose) for labeling free or protein-conjugated ADP-ribose monomers and polymers at their 2'-OH termini using the enzyme OAS1 and dATP. When coupled with various dATP analogs (e.g., radioactive, fluorescent, affinity tags), ELTA can be used to explore PAR biology with techniques routinely used to investigate DNA or RNA function. We demonstrate that ELTA enables the biophysical measurements of protein binding to PAR of a defined length, detection of PAR length from proteins and cells, and enrichment of sub-femtomole amounts of ADP-ribosylated peptides from cell lysates.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , ADP-Ribosylation , Adenosine Diphosphate Ribose/metabolism , Deoxyadenine Nucleotides/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Ubiquitin-Protein Ligases/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , Animals , HeLa Cells , Humans , Protein Binding , Protein Domains , Sf9 Cells , Ubiquitin-Protein Ligases/genetics
8.
Proc Natl Acad Sci U S A ; 121(21): e2322974121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38743621

ABSTRACT

SRSF1 is the founding member of the SR protein family. It is required-interchangeably with other SR proteins-for pre-mRNA splicing in vitro, and it regulates various alternative splicing events. Dysregulation of SRSF1 expression contributes to cancer and other pathologies. Here, we characterized SRSF1's interactome using proximity labeling and mass spectrometry. This approach yielded 190 proteins enriched in the SRSF1 samples, independently of the N- or C-terminal location of the biotin-labeling domain. The detected proteins reflect established functions of SRSF1 in pre-mRNA splicing and reveal additional connections to spliceosome proteins, in addition to other recently identified functions. We validated a robust interaction with the spliceosomal RNA helicase DDX23/PRP28 using bimolecular fluorescence complementation and in vitro binding assays. The interaction is mediated by the N-terminal RS-like domain of DDX23 and both RRM1 and the RS domain of SRSF1. During pre-mRNA splicing, DDX23's ATPase activity is essential for the pre-B to B spliceosome complex transition and for release of U1 snRNP from the 5' splice site. We show that the RS-like region of DDX23's N-terminal domain is important for spliceosome incorporation, while larger deletions in this domain alter subnuclear localization. We discuss how the identified interaction of DDX23 with SRSF1 and other SR proteins may be involved in the regulation of these processes.


Subject(s)
DEAD-box RNA Helicases , Serine-Arginine Splicing Factors , Spliceosomes , Humans , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , HeLa Cells , Protein Binding , RNA Precursors/metabolism , RNA Precursors/genetics , RNA Splicing , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Spliceosomes/metabolism
9.
Mol Cell ; 69(5): 787-801.e8, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29499134

ABSTRACT

MicroRNA-mediated gene silencing is a fundamental mechanism in the regulation of gene expression. It remains unclear how the efficiency of RNA silencing could be influenced by RNA-binding proteins associated with the microRNA-induced silencing complex (miRISC). Here we report that fused in sarcoma (FUS), an RNA-binding protein linked to neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), interacts with the core miRISC component AGO2 and is required for optimal microRNA-mediated gene silencing. FUS promotes gene silencing by binding to microRNA and mRNA targets, as illustrated by its action on miR-200c and its target ZEB1. A truncated mutant form of FUS that leads its carriers to an aggressive form of ALS, R495X, impairs microRNA-mediated gene silencing. The C. elegans homolog fust-1 also shares a conserved role in regulating the microRNA pathway. Collectively, our results suggest a role for FUS in regulating the activity of microRNA-mediated silencing.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Gene Silencing , MicroRNAs/metabolism , RNA, Helminth/metabolism , RNA-Binding Protein FUS/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , HEK293 Cells , Humans , Mice , MicroRNAs/genetics , RNA, Helminth/genetics , RNA-Binding Protein FUS/genetics
10.
Nucleic Acids Res ; 52(9): 4985-5001, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38471816

ABSTRACT

Many microRNA (miRNA)-guided Argonaute proteins can cleave RNA ('slicing'), even though miRNA-mediated target repression is generally cleavage-independent. Here we use Caenorhabditis elegans to examine the role of catalytic residues of miRNA Argonautes in organismal development. In contrast to previous work, mutations in presumed catalytic residues did not interfere with development when introduced by CRISPR. We find that unwinding and decay of miRNA star strands is weakly defective in the catalytic residue mutants, with the largest effect observed in embryos. Argonaute-Like Gene 2 (ALG-2) is more dependent on catalytic residues for unwinding than ALG-1. The miRNAs that displayed the greatest (albeit minor) dependence on catalytic residues for unwinding tend to form stable duplexes with their star strand, and in some cases, lowering duplex stability alleviates dependence on catalytic residues. While a few miRNA guide strands are reduced in the mutant background, the basis of this is unclear since changes were not dependent on EBAX-1, an effector of Target-Directed miRNA Degradation (TDMD). Overall, this work defines a role for the catalytic residues of miRNA Argonautes in star strand decay; future work should examine whether this role contributes to the selection pressure to conserve catalytic activity of miRNA Argonautes across the metazoan phylogeny.


Subject(s)
Argonaute Proteins , Caenorhabditis elegans Proteins , Caenorhabditis elegans , MicroRNAs , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/chemistry , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/chemistry , RNA Stability/genetics , Mutation , Catalytic Domain/genetics , CRISPR-Cas Systems , RNA-Binding Proteins
11.
Mol Cell ; 67(4): 646-658.e3, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28781232

ABSTRACT

In miRNA-mediated gene silencing, the physical interaction between human Argonaute (hAgo) and GW182 (hGW182) is essential for facilitating the downstream silencing of the targeted mRNA. GW182 can interact with hAgo via three of the GW/WG repeats in its Argonaute-binding domain: motif-1, motif-2, and the hook motif. The structure of hAgo1 in complex with the hook motif of hGW182 reveals a "gate"-like interaction that is critical for GW182 docking into one of hAgo1's tryptophan-binding pockets. We show that hAgo1 and hAgo2 have a single GW182-binding site and that miRNA binding increases hAgo's affinity to GW182. With target binding occurring rapidly, this ensures that only mature RISC would be recruited for silencing. Finally, we show that hGW182 can recruit up to three copies of hAgo via its three GW motifs. This may explain the observed cooperativity in miRNA-mediated gene silencing.


Subject(s)
Argonaute Proteins/metabolism , Autoantigens/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Silencing , MicroRNAs/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA-Binding Proteins/metabolism , Animals , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Autoantigens/chemistry , Autoantigens/genetics , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/genetics , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , Molecular Docking Simulation , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Sf9 Cells , Structure-Activity Relationship , Transfection
12.
Mol Cell ; 60(6): 847-59, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26626481

ABSTRACT

The bromodomain and extraterminal (BET) protein BRD4 is a therapeutic target in acute myeloid leukemia (AML). Here, we demonstrate that the AML maintenance function of BRD4 requires its interaction with NSD3, which belongs to a subfamily of H3K36 methyltransferases. Unexpectedly, AML cells were found to only require a short isoform of NSD3 that lacks the methyltransferase domain. We show that NSD3-short is an adaptor protein that sustains leukemia by linking BRD4 to the CHD8 chromatin remodeler, by using a PWWP chromatin reader module, and by employing an acidic transactivation domain. Genetic targeting of NSD3 or CHD8 mimics the phenotypic and transcriptional effects of BRD4 inhibition. Furthermore, BRD4, NSD3, and CHD8 colocalize across the AML genome, and each is released from super-enhancer regions upon chemical inhibition of BET bromodomains. These findings suggest that BET inhibitors exert therapeutic effects in leukemia by evicting BRD4-NSD3-CHD8 complexes from chromatin to suppress transcription.


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Catalytic Domain , Cell Cycle Proteins , Cell Proliferation , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , HL-60 Cells , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/genetics
13.
Mol Cell ; 58(2): 255-68, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25866245

ABSTRACT

PTEN is proposed to function at the plasma membrane, where receptor tyrosine kinases are activated. However, the majority of PTEN is located throughout the cytoplasm. Here, we show that cytoplasmic PTEN is distributed along microtubules, tethered to vesicles via phosphatidylinositol 3-phosphate (PI(3)P), the signature lipid of endosomes. We demonstrate that the non-catalytic C2 domain of PTEN specifically binds PI(3)P through the CBR3 loop. Mutations render this loop incapable of PI(3)P binding and abrogate PTEN-mediated inhibition of PI 3-kinase/AKT signaling. This loss of function is rescued by fusion of the loop mutant PTEN to FYVE, the canonical PI(3)P binding domain, demonstrating the functional importance of targeting PTEN to endosomal membranes. Beyond revealing an upstream activation mechanism of PTEN, our data introduce the concept of PI 3-kinase signal activation on the vast plasma membrane that is contrasted by PTEN-mediated signal termination on the small, discrete surfaces of internalized vesicles.


Subject(s)
PTEN Phosphohydrolase/chemistry , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Transport Vesicles/metabolism , Animals , Binding Sites , Mice , Microtubules/enzymology , Models, Molecular , NIH 3T3 Cells , Protein Structure, Secondary , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 116(26): 12828-12832, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31182604

ABSTRACT

Noroviruses are a leading cause of foodborne illnesses worldwide. Although GII.4 strains have been responsible for most norovirus outbreaks, the assembled virus shell structures have been available in detail for only a single strain (GI.1). We present high-resolution (2.6- to 4.1-Å) cryoelectron microscopy (cryo-EM) structures of GII.4, GII.2, GI.7, and GI.1 human norovirus outbreak strain virus-like particles (VLPs). Although norovirus VLPs have been thought to exist in a single-sized assembly, our structures reveal polymorphism between and within genogroups, with small, medium, and large particle sizes observed. Using asymmetric reconstruction, we were able to resolve a Zn2+ metal ion adjacent to the coreceptor binding site, which affected the structural stability of the shell. Our structures serve as valuable templates for facilitating vaccine formulations.


Subject(s)
Capsid/ultrastructure , Disease Outbreaks , Norovirus/ultrastructure , Caliciviridae Infections/virology , Capsid/metabolism , Cryoelectron Microscopy , Genetic Variation , Humans , Norovirus/genetics , Norovirus/isolation & purification , Protein Binding , Zinc/metabolism
15.
Proc Natl Acad Sci U S A ; 115(12): E2696-E2705, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29432194

ABSTRACT

Small interfering RNA (siRNA) represents a promising class of inhibitors in both fundamental research and the clinic. Numerous delivery vehicles have been developed to facilitate siRNA delivery. Nevertheless, achieving highly potent RNA interference (RNAi) toward clinical translation requires efficient formation of RNA-induced gene-silencing complex (RISC) in the cytoplasm. Here we coencapsulate siRNA and the central RNAi effector protein Argonaute 2 (Ago2) via different delivery carriers as a platform to augment RNAi. The physical clustering between siRNA and Ago2 is found to be indispensable for enhanced RNAi. Moreover, by utilizing polyamines bearing the same backbone but distinct cationic side-group arrangements of ethylene diamine repeats as the delivery vehicles, we find that the molecular structure of these polyamines modulates the degree of siRNA/Ago2-mediated improvement of RNAi. We apply this strategy to silence the oncogene STAT3 and significantly prolong survival in mice challenged with melanoma. Our findings suggest a paradigm for RNAi via the synergistic coassembly of RNA with helper proteins.


Subject(s)
Argonaute Proteins/genetics , Genetic Therapy/methods , RNA Interference , RNA, Small Interfering/administration & dosage , RNA-Induced Silencing Complex/chemistry , Animals , Argonaute Proteins/metabolism , Drug Delivery Systems/methods , Melanoma, Experimental/genetics , Melanoma, Experimental/mortality , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Oncogenes/genetics , Polyamines/chemistry , RNA, Antisense/administration & dosage , RNA, Antisense/pharmacology , RNA, Double-Stranded/administration & dosage , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Messenger , RNA, Small Interfering/chemistry , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , STAT3 Transcription Factor/genetics , Structure-Activity Relationship , Transfection/methods
16.
Nature ; 514(7521): 252-256, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25119025

ABSTRACT

The pluripotency factor Lin28 inhibits the biogenesis of the let-7 family of mammalian microRNAs. Lin28 is highly expressed in embryonic stem cells and has a fundamental role in regulation of development, glucose metabolism and tissue regeneration. Overexpression of Lin28 is correlated with the onset of numerous cancers, whereas let-7, a tumour suppressor, silences several human oncogenes. Lin28 binds to precursor let-7 (pre-let-7) hairpins, triggering the 3' oligo-uridylation activity of TUT4 and TUT7 (refs 10-12). The oligoU tail added to pre-let-7 serves as a decay signal, as it is rapidly degraded by Dis3l2 (refs 13, 14), a homologue of the catalytic subunit of the RNA exosome. The molecular basis of Lin28-mediated recruitment of TUT4 and TUT7 to pre-let-7 and its subsequent degradation by Dis3l2 is largely unknown. To examine the mechanism of Dis3l2 substrate recognition we determined the structure of mouse Dis3l2 in complex with an oligoU RNA to mimic the uridylated tail of pre-let-7. Three RNA-binding domains form an open funnel on one face of the catalytic domain that allows RNA to navigate a path to the active site different from that of its exosome counterpart. The resulting path reveals an extensive network of uracil-specific interactions spanning the first 12 nucleotides of an oligoU-tailed RNA. We identify three U-specificity zones that explain how Dis3l2 recognizes, binds and processes uridylated pre-let-7 in the final step of the Lin28-let-7 pathway.


Subject(s)
Exoribonucleases/chemistry , Exoribonucleases/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Animals , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Exosome Multienzyme Ribonuclease Complex/chemistry , Mice , MicroRNAs/chemistry , MicroRNAs/genetics , Models, Molecular , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism , RNA-Binding Proteins/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Substrate Specificity , Uracil Nucleotides/chemistry , Uracil Nucleotides/metabolism
17.
Genes Dev ; 26(3): 294-303, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22302941

ABSTRACT

A wealth of genetic information and some biochemical analysis have made the GAL regulon of the yeast Saccharomyces cerevisiae a classic model system for studying transcriptional activation in eukaryotes. Galactose induces this transcriptional switch, which is regulated by three proteins: the transcriptional activator Gal4p, bound to DNA; the repressor Gal80p; and the transducer Gal3p. We showed previously that NADP appears to act as a trigger to kick the repressor off the activator. Sustained activation involves a complex of the transducer Gal3p and Gal80p mediated by galactose and ATP. We solved the crystal structure of the complex of Gal3p-Gal80p with α-D-galactose and ATP to 2.1 Å resolution. The interaction between the proteins occurs only when Gal3p is in a "closed" state induced by ligand binding. The structure of the complex provides a rationale for the phenotypes of several well-known Gal80p and Gal3p mutants as well as the lack of galactokinase activity of Gal3p.


Subject(s)
Ligands , Models, Molecular , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Galactokinase/chemistry , Galactose/chemistry , Galactose/metabolism , Humans , Hydrogen Bonding , Mutation , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Regulon , Repressor Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcriptional Activation
18.
Genes Dev ; 26(21): 2392-407, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23073843

ABSTRACT

The MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) locus is misregulated in many human cancers and produces an abundant long nuclear-retained noncoding RNA. Despite being transcribed by RNA polymerase II, the 3' end of MALAT1 is produced not by canonical cleavage/polyadenylation but instead by recognition and cleavage of a tRNA-like structure by RNase P. Mature MALAT1 thus lacks a poly(A) tail yet is expressed at a level higher than many protein-coding genes in vivo. Here we show that the 3' ends of MALAT1 and the MEN ß long noncoding RNAs are protected from 3'-5' exonucleases by highly conserved triple helical structures. Surprisingly, when these structures are placed downstream from an ORF, the transcript is efficiently translated in vivo despite the lack of a poly(A) tail. The triple helix therefore also functions as a translational enhancer, and mutations in this region separate this translation activity from simple effects on RNA stability or transport. We further found that a transcript ending in a triple helix is efficiently repressed by microRNAs in vivo, arguing against a major role for the poly(A) tail in microRNA-mediated silencing. These results provide new insights into how transcripts that lack poly(A) tails are stabilized and regulated and suggest that RNA triple-helical structures likely have key regulatory functions in vivo.


Subject(s)
RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Amino Acid Motifs , Base Sequence , DNA Mutational Analysis , Gene Expression Regulation , HeLa Cells , Humans , MicroRNAs/metabolism , Molecular Sequence Data , Plasmids/genetics , Protein Denaturation , Protein Structure, Secondary , RNA 3' End Processing/genetics , RNA Stability , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/metabolism , Sequence Alignment
19.
Nucleic Acids Res ; 45(6): 3528-3536, 2017 04 07.
Article in English | MEDLINE | ID: mdl-27903888

ABSTRACT

Efficient gene silencing by RNA interference (RNAi) in vivo requires the recognition and binding of the 5΄- phosphate of the guide strand of an siRNA by the Argonaute protein. However, for exogenous siRNAs it is limited by the rapid removal of the 5΄- phosphate of the guide strand by metabolic enzymes. Here, we have determined the crystal structure of human Argonaute-2 in complex with the metabolically stable 5΄-(E)-vinylphosphonate (5΄-E-VP) guide RNA at 2.5-Šresolution. The structure demonstrates how the 5΄ binding site in the Mid domain of human Argonaute-2 is able to adjust the key residues in the 5΄-nucleotide binding pocket to compensate for the change introduced by the modified nucleotide. This observation also explains improved binding affinity of the 5΄-E-VP -modified siRNA to human Argonaute-2 in-vitro, as well as the enhanced silencing in the context of the trivalent N-acetylgalactosamine (GalNAc)-conjugated siRNA in mice relative to the un-modified siRNA.


Subject(s)
Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Organophosphonates/chemistry , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Vinyl Compounds/chemistry , Animals , Binding Sites , Humans , Mice , Models, Molecular , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/metabolism , Receptors, Albumin/genetics , Receptors, Albumin/metabolism
20.
Nature ; 491(7423): 279-83, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23064227

ABSTRACT

PIWI-family proteins and their associated small RNAs (piRNAs) act in an evolutionarily conserved innate immune mechanism to provide essential protection for germ-cell genomes against the activity of mobile genetic elements. piRNA populations comprise a molecular definition of transposons, which permits them to distinguish transposons from host genes and selectively silence them. piRNAs can be generated in two distinct ways, forming either primary or secondary piRNAs. Primary piRNAs come from discrete genomic loci, termed piRNA clusters, and seem to be derived from long, single-stranded precursors. The biogenesis of primary piRNAs involves at least two nucleolytic steps. An unknown enzyme cleaves piRNA cluster transcripts to generate monophosphorylated piRNA 5' ends. piRNA 3' ends are probably formed by exonucleolytic trimming, after a piRNA precursor is loaded into its PIWI partner. Secondary piRNAs arise during the adaptive 'ping-pong' cycle, with their 5' termini being formed by the activity of PIWIs themselves. A number of proteins have been implicated genetically in primary piRNA biogenesis. One of these, Drosophila melanogaster Zucchini, is a member of the phospholipase-D family of phosphodiesterases, which includes both phospholipases and nucleases. Here we produced a dimeric, soluble fragment of the mouse Zucchini homologue (mZuc; also known as PLD6) and show that it possesses single-strand-specific nuclease activity. A crystal structure of mZuc at 1.75 Å resolution indicates greater architectural similarity to phospholipase-D family nucleases than to phospholipases. Together, our data suggest that the Zucchini proteins act in primary piRNA biogenesis as nucleases, perhaps generating the 5' ends of primary piRNAs.


Subject(s)
Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Phospholipase D/chemistry , Phospholipase D/metabolism , RNA, Small Interfering/metabolism , Animals , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Mice , Models, Molecular , Protein Conformation , Protein Multimerization , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Static Electricity , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL