Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 31(5): 754-771.e6, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701759

ABSTRACT

Development of embryonic stem cells (ESCs) into neurons requires intricate regulation of transcription, splicing, and translation, but how these processes interconnect is not understood. We found that polypyrimidine tract binding protein 1 (PTBP1) controls splicing of DPF2, a subunit of BRG1/BRM-associated factor (BAF) chromatin remodeling complexes. Dpf2 exon 7 splicing is inhibited by PTBP1 to produce the DPF2-S isoform early in development. During neuronal differentiation, loss of PTBP1 allows exon 7 inclusion and DPF2-L expression. Different cellular phenotypes and gene expression programs were induced by these alternative DPF2 isoforms. We identified chromatin binding sites enriched for each DPF2 isoform, as well as sites bound by both. In ESC, DPF2-S preferential sites were bound by pluripotency factors. In neuronal progenitors, DPF2-S sites were bound by nuclear factor I (NFI), while DPF2-L sites were bound by CCCTC-binding factor (CTCF). DPF2-S sites exhibited enhancer modifications, while DPF2-L sites showed promoter modifications. Thus, alternative splicing redirects BAF complex targeting to impact chromatin organization during neuronal development.


Subject(s)
Alternative Splicing , Cell Differentiation , Chromatin , Heterogeneous-Nuclear Ribonucleoproteins , Neurons , Polypyrimidine Tract-Binding Protein , Transcription Factors , Alternative Splicing/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Animals , Cell Differentiation/genetics , Chromatin/metabolism , Mice , Neurons/metabolism , Neurons/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription, Genetic , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Exons/genetics , Humans , Cell Self Renewal/genetics
2.
Genes Dev ; 38(7-8): 294-307, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38688681

ABSTRACT

Synaptic function in neurons is modulated by local translation of mRNAs that are transported to distal portions of axons and dendrites. The metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is broadly expressed across cell types, almost exclusively as a nuclear long noncoding RNA. We found that in differentiating neurons, a portion of Malat1 RNA redistributes to the cytoplasm. Depletion of Malat1 using antisense oligonucleotides (ASOs) stimulates the expression of particular pre- and postsynaptic proteins, implicating Malat1 in their regulation. Neuronal Malat1 is localized in puncta of both axons and dendrites that costain with Staufen1 protein, similar to neuronal RNA granules formed by locally translated mRNAs. Ribosome profiling of cultured mouse cortical neurons identified ribosome footprints within a 5' region of Malat1 containing short open reading frames. The upstream-most reading frame (M1) of the Malat1 locus was linked to the GFP-coding sequence in mouse embryonic stem cells. When these gene-edited cells were differentiated into glutamatergic neurons, the M1-GFP fusion protein was expressed. Antibody staining for the M1 peptide confirmed its presence in wild-type neurons and showed that M1 expression was enhanced by synaptic stimulation with KCl. Our results indicate that Malat1 serves as a cytoplasmic coding RNA in the brain that is both modulated by and modulates synaptic function.


Subject(s)
Cytoplasm , Neurons , RNA, Long Noncoding , RNA, Messenger , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice , Neurons/metabolism , Cytoplasm/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cells, Cultured , Cell Differentiation , Peptides/metabolism , Peptides/genetics
3.
bioRxiv ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38352368

ABSTRACT

Synaptic function is modulated by local translation of mRNAs that are transported to distal portions of axons and dendrites. The Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is broadly expressed across cell types, almost exclusively as a nuclear non-coding RNA. We found that in differentiating neurons, a portion of Malat1 RNA redistributes to the cytoplasm. Depletion of Malat1 from neurons stimulated expression of particular pre- and post- synaptic proteins, implicating Malat1 in their regulation. Neuronal Malat1 is localized to both axons and dendrites in puncta that co-stain with Staufen1 protein, similar to neuronal granules formed by locally translated mRNAs. Ribosome profiling of mouse cortical neurons identified ribosome footprints within a region of Malat1 containing short open reading frames. The upstream-most reading frame (M1) of the Malat1 locus was linked to the GFP coding sequence in mouse ES cells. When these gene-edited cells were differentiated into glutamatergic neurons, the M1-GFP fusion protein was expressed. Antibody staining for the M1 peptide confirmed its presence in wildtype neurons, and showed enhancement of M1 expression after synaptic stimulation with KCL. Our results indicate that Malat1 serves as a cytoplasmic coding RNA in the brain that is both modulated by and modulates synaptic function.

4.
RNA ; 29(8): 1274-1287, 2023 08.
Article in English | MEDLINE | ID: mdl-37130703

ABSTRACT

Fluorescence in situ hybridization (FISH) is a widely used tool for quantifying gene expression and determining the location of RNA molecules in cells. We present an improved method for FISH probe production that yields high-purity probes with a wide range of fluorophores using standard laboratory equipment at low cost. The method modifies an earlier protocol that uses terminal deoxynucleotidyl transferase to add fluorescently labeled nucleotides to synthetic deoxyoligonucleotides. In our protocol, amino-11-ddUTP is joined to an oligonucleotide pool prior to its conjugation to a fluorescent dye, thereby generating pools of probes ready for a variety of modifications. This order of reaction steps allows for high labeling efficiencies regardless of the GC content or terminal base of the oligonucleotides. The degree of labeling (DOL) for spectrally distinct fluorophores (Quasar, ATTO, and Alexa dyes) was mostly >90%, comparable with commercial probes. The ease and low cost of production allowed the generation of probe sets targeting a wide variety of RNA molecules. Using these probes, FISH assays in C2C12 cells showed the expected subcellular localization of mRNAs and pre-mRNAs for Polr2a (RNA polymerase II subunit 2a) and Gapdh, and of the long noncoding RNAs Malat1 and Neat1 Developing FISH probe sets for several transcripts containing retained introns, we found that retained introns in the Gabbr1 and Noc2l transcripts are present in subnuclear foci separate from their sites of synthesis and partially coincident with nuclear speckles. This labeling protocol should have many applications in RNA biology.


Subject(s)
Oligonucleotides , RNA , In Situ Hybridization, Fluorescence/methods , Introns/genetics , RNA, Messenger/genetics , Oligonucleotide Probes/genetics , Fluorescent Dyes
5.
Genome Res ; 31(6): 1106-1119, 2021 06.
Article in English | MEDLINE | ID: mdl-33832989

ABSTRACT

Steps of mRNA maturation are important gene regulatory events that occur in distinct cellular locations. However, transcriptomic analyses often lose information on the subcellular distribution of processed and unprocessed transcripts. We generated extensive RNA-seq data sets to track mRNA maturation across subcellular locations in mouse embryonic stem cells, neuronal progenitor cells, and postmitotic neurons. We find disparate patterns of RNA enrichment between the cytoplasmic, nucleoplasmic, and chromatin fractions, with some genes maintaining more polyadenylated RNA in chromatin than in the cytoplasm. We bioinformatically defined four regulatory groups for intron retention, including complete cotranscriptional splicing, complete intron retention in the cytoplasmic RNA, and two intron groups present in nuclear and chromatin transcripts but fully excised in cytoplasm. We found that introns switch their regulatory group between cell types, including neuronally excised introns repressed by polypyrimidine track binding protein 1 (PTBP1). Transcripts for the neuronal gamma-aminobutyric acid (GABA) B receptor, 1 (Gabbr1) are highly expressed in mESCs but are absent from the cytoplasm. Instead, incompletely spliced Gabbr1 RNA remains sequestered on chromatin, where it is bound by PTBP1, similar to certain long noncoding RNAs. Upon neuronal differentiation, Gabbr1 RNA becomes fully processed and exported for translation. Thus, splicing repression and chromatin anchoring of RNA combine to allow posttranscriptional regulation of Gabbr1 over development. For this and other genes, polyadenylated RNA abundance does not indicate functional gene expression. Our data sets provide a rich resource for analyzing many other aspects of mRNA maturation in subcellular locations and across development.


Subject(s)
RNA Precursors , RNA Splicing , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Genes, Developmental , Introns/genetics , Mice , RNA Precursors/genetics , RNA Precursors/metabolism
6.
Proc Natl Acad Sci U S A ; 115(47): E11061-E11070, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30401736

ABSTRACT

MicroRNA (miRNA)-124 is expressed in neurons, where it represses genes inhibitory for neuronal differentiation, including the RNA binding protein PTBP1. PTBP1 maintains nonneuronal splicing patterns of mRNAs that switch to neuronal isoforms upon neuronal differentiation. We find that primary (pri)-miR-124-1 is expressed in mouse embryonic stem cells where mature miR-124 is absent. PTBP1 binds to this precursor RNA upstream of the miRNA stem-loop to inhibit mature miR-124 expression in vivo and DROSHA cleavage of pri-miR-124-1 in vitro. This function for PTBP1 in repressing miR-124 biogenesis defines an additional regulatory loop in the already intricate interplay between these two molecules. Applying mathematical modeling to examine the dynamics of this regulation, we find that the pool of pri-miR-124 whose maturation is blocked by PTBP1 creates a robust and self-reinforcing transition in gene expression as PTBP1 is depleted during early neuronal differentiation. While interlocking regulatory loops are often found between miRNAs and transcriptional regulators, our results indicate that miRNA targeting of posttranscriptional regulators also reinforces developmental decisions. Notably, induction of neuronal differentiation observed upon PTBP1 knockdown likely results from direct derepression of miR-124, in addition to indirect effects previously described.


Subject(s)
Gene Expression Regulation/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neurons/cytology , Polypyrimidine Tract-Binding Protein/metabolism , Animals , Cell Line, Tumor , Embryonic Stem Cells/metabolism , Gene Knockout Techniques , Mice , Models, Theoretical , Neuroblastoma/metabolism , Neurogenesis/genetics , RNA Processing, Post-Transcriptional/genetics , Ribonuclease III/metabolism
7.
Methods Mol Biol ; 1648: 155-167, 2017.
Article in English | MEDLINE | ID: mdl-28766296

ABSTRACT

Eukaryotic organelles or subcellular compartments can be selectively isolated based on their physical density and their stability in the presence of nonionic detergents. This chapter describes a protocol for the preparation of cytoplasm, nucleoplasm, and chromatin, in addition to isolation of RNA and proteins from these fractions. Proteins and protein complexes stably associated with chromatin and other high molecular weight nuclear components can be extracted under non-denaturing conditions by enzymatic digestion of RNA and DNA. The chapter also includes a detailed extraction protocol from highly purified nuclei.


Subject(s)
Chromatin/chemistry , Cytoplasm/chemistry , Multiprotein Complexes , RNA , Animals , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/isolation & purification , RNA/chemistry , RNA/isolation & purification
8.
Nat Commun ; 7: 13694, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27934859

ABSTRACT

The RNA-binding protein TRBP is a central component of the Dicer complex. Despite a decade of biochemical and structural studies, the essential functionality of TRBP in microRNA (miRNA) biogenesis remains unknown. Here we show that TRBP is an integral cofactor for time-efficient Dicer processing in RNA-crowded environments. We competed for Dicer processing of pre-miRNA with a large amount of cellular RNA species and found that Dicer-TRBP, but not Dicer alone, remains resilient. To apprehend the mechanism of this substrate selectivity, we use single-molecule fluorescence. The real-time observation reveals that TRBP acts as a gatekeeper, precluding Dicer from engaging with pre-miRNA-like substrates. TRBP acquires the selectivity using the PAZ domain of Dicer, whereas Dicer moderates the RNA-binding affinity of TRBP for fast turnover. This coordinated action between TRBP and Dicer accomplishes an efficient way of discarding pre-miRNA-like substrates.


Subject(s)
MicroRNAs/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Nuclear Receptor Coactivators , Protein Binding , Protein Domains , RNA , RNA-Binding Proteins/genetics , Transcription, Genetic
9.
RNA ; 22(8): 1172-80, 2016 08.
Article in English | MEDLINE | ID: mdl-27288314

ABSTRACT

Most human genes generate multiple protein isoforms through alternative pre-mRNA splicing, but the mechanisms controlling alternative splicing choices by RNA binding proteins are not well understood. These proteins can have multiple paralogs expressed in different cell types and exhibiting different splicing activities on target exons. We examined the paralogous polypyrimidine tract binding proteins PTBP1 and PTBP2 to understand how PTBP1 can exhibit greater splicing repression activity on certain exons. Using both an in vivo coexpression assay and an in vitro splicing assay, we show that PTBP1 is more repressive than PTBP2 per unit protein on a target exon. Constructing chimeras of PTBP1 and 2 to determine amino acid features that contribute to their differential activity, we find that multiple segments of PTBP1 increase the repressive activity of PTBP2. Notably, when either RRM1 of PTBP2 or the linker peptide separating RRM2 and RRM3 are replaced with the equivalent PTBP1 sequences, the resulting chimeras are highly active for splicing repression. These segments are distinct from the known region of interaction for the PTBP1 cofactors Raver1 and Matrin3 in RRM2. We find that RRM2 of PTBP1 also increases the repression activity of an otherwise PTBP2 sequence, and that this is potentially explained by stronger binding by Raver1. These results indicate that multiple features over the length of the two proteins affect their ability to repress an exon.


Subject(s)
Polypyrimidine Tract-Binding Protein/metabolism , RNA Splicing , Animals , Cells, Cultured , Exons , Humans , Polypyrimidine Tract-Binding Protein/chemistry , Protein Conformation
10.
Methods ; 105: 99-108, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27017911

ABSTRACT

The genome and transcriptome are constantly modified by proteins in the cell. Recent advances in single-molecule techniques allow for high spatial and temporal observations of these interactions between proteins and nucleic acids. However, due to the difficulty of obtaining functional protein complexes, it remains challenging to study the interactions between macromolecular protein complexes and nucleic acids. Here, we combined single-molecule fluorescence with various protein complex pull-down techniques to determine the function and stoichiometry of ribonucleoprotein complexes. Through the use of three examples of protein complexes from eukaryotic cells (Drosha, Dicer, and TUT4 protein complexes), we provide step-by-step guidance for using novel single-molecule techniques. Our single-molecule methods provide sub-second and nanometer resolution and can be applied to other nucleoprotein complexes that are essential for cellular processes.


Subject(s)
DNA-Binding Proteins/chemistry , Microscopy, Fluorescence/methods , Multiprotein Complexes/chemistry , Single Molecule Imaging/methods , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , Humans , Multiprotein Complexes/genetics , Nanotechnology/methods , Nucleic Acids/chemistry , Nucleic Acids/genetics , Ribonuclease III/chemistry , Ribonuclease III/genetics
11.
EMBO Rep ; 12(7): 690-6, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21637296

ABSTRACT

Single-molecule techniques have been used for only a subset of biological problems because of difficulties in studying proteins that require cofactors or post-translational modifications. Here, we present a new method integrating single-molecule fluorescence microscopy and immunopurification to study protein complexes. We used this method to investigate Lin28-mediated microRNA uridylation by TUT4 (terminal uridylyl transferase 4, polyU polymerase), which regulates let-7 microRNA biogenesis. Our real-time analysis of the uridylation by the TUT4 immunoprecipitates suggests that Lin28 functions as a processivity factor of TUT4. Our new technique, SIMPlex (single-molecule approach to immunoprecipitated protein complexes), provides a universal tool to analyse complex proteins at the single-molecule level.


Subject(s)
Biological Assay , Immunoprecipitation/methods , MicroRNAs/metabolism , Multiprotein Complexes/metabolism , RNA Nucleotidyltransferases/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Protein Binding , RNA-Binding Proteins
12.
Mol Cell Biol ; 29(21): 5789-99, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19704008

ABSTRACT

MicroRNAs (miRNAs) are endogenous antisense regulators that trigger endonucleolytic mRNA cleavage, translational repression, and/or mRNA decay. miRNA-mediated gene regulation is important for numerous biological pathways, yet the underlying mechanisms are still under rigorous investigation. Here we identify human UPF1 (hUPF1) as a protein that contributes to RNA silencing. When hUPF1 is knocked down, miRNA targets are upregulated. The depletion of hUPF1 also increases the off-target messages of small interfering RNAs (siRNAs), which are imperfectly complementary to transfected siRNAs. Conversely, when overexpressed, wild-type hUPF1 downregulates miRNA targets. The helicase domain mutant of hUPF1 fails to suppress miRNA targets. hUPF1 interacts with human Argonaute 1 (hAGO1) and hAGO2 and colocalizes with hAGO1 and hAGO2 in processing bodies, which are known to be the sites for translational repression and mRNA destruction. We further find that the amounts of target messages bound to hAGO2 are reduced when hUPF1 is depleted. Our data thus suggest that hUPF1 may participate in RNA silencing by facilitating the binding of the RNA-induced silencing complex to the target and by accelerating the decay of the mRNA.


Subject(s)
Down-Regulation/genetics , MicroRNAs/metabolism , RNA, Small Interfering/metabolism , Trans-Activators/metabolism , Argonaute Proteins , Cytoplasmic Structures/metabolism , Eukaryotic Initiation Factor-2/metabolism , Gene Knockdown Techniques , Gene Silencing , HeLa Cells , Humans , Models, Biological , Molecular Mimicry , Protein Binding , Protein Transport , RNA Helicases , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/metabolism , Subcellular Fractions/metabolism
13.
Cell ; 138(4): 696-708, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19703396

ABSTRACT

As key regulators in cellular functions, microRNAs (miRNAs) themselves need to be tightly controlled. Lin28, a pluripotency factor, was reported to downregulate let-7 miRNA by inducing uridylation of let-7 precursor (pre-let-7). But the enzyme responsible for the uridylation remained unknown. Here we identify a noncanonical poly (A) polymerase, TUTase4 (TUT4), as the uridylyl transferase for pre-let-7. Lin28 recruits TUT4 to pre-let-7 by recognizing a tetra-nucleotide sequence motif (GGAG) in the terminal loop. TUT4 in turn adds an oligouridine tail to the pre-let-7, which blocks Dicer processing. Other miRNAs with the same sequence motif (miR-107, -143, and -200c) are regulated through the same mechanism. Knockdown of TUT4 and Lin28 reduces the level of stem cell markers, suggesting that they are required for stem cell maintenance. This study uncovers the role of TUT4 and Lin28 as specific suppressors of miRNA biogenesis, which has implications for stem cell research and cancer biology.


Subject(s)
Embryonic Stem Cells/cytology , MicroRNAs/metabolism , Polynucleotide Adenylyltransferase/metabolism , Uridine/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Humans , Mice
14.
Cell ; 136(1): 75-84, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19135890

ABSTRACT

The Drosha-DGCR8 complex, also known as Microprocessor, is essential for microRNA (miRNA) maturation. Drosha functions as the catalytic subunit, while DGCR8 (also known as Pasha) recognizes the RNA substrate. Although the action mechanism of this complex has been intensively studied, it remains unclear how Drosha and DGCR8 are regulated and if these proteins have any additional role(s) apart from miRNA processing. Here, we report that Drosha and DGCR8 regulate each other posttranscriptionally. The Drosha-DGCR8 complex cleaves the hairpin structures embedded in the DGCR8 mRNA and thereby destabilizes the mRNA. We further find that DGCR8 stabilizes the Drosha protein via protein-protein interaction. This crossregulation between Drosha and DGCR8 may contribute to the homeostatic control of miRNA biogenesis. Furthermore, microarray analyses suggest that a number of mRNAs may be downregulated in a Microprocessor-dependent, miRNA-independent manner. Our study reveals a previously unsuspected function of Microprocessor in mRNA stability control.


Subject(s)
Gene Expression Regulation , Proteins/genetics , RNA Stability , Ribonuclease III/genetics , Animals , Base Sequence , Cell Line , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Proteins/metabolism , RNA Interference , RNA-Binding Proteins , Ribonuclease III/metabolism
15.
Nat Struct Mol Biol ; 14(9): 847-53, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17704815

ABSTRACT

A complex of Drosha with DGCR8 (or its homolog Pasha) cleaves primary microRNA (pri-miRNA) substrates into precursor miRNA and initiates the microRNA maturation process. Drosha provides the catalytic site for this cleavage, whereas DGCR8 or Pasha provides a frame for anchoring substrate pri-miRNAs. To clarify the molecular basis underlying recognition of pri-miRNA by DGCR8 and Pasha, we determined the crystal structure of the human DGCR8 core (DGCR8S, residues 493-720). In the structure, the two double-stranded RNA-binding domains (dsRBDs) are arranged with pseudo two-fold symmetry and are tightly packed against the C-terminal helix. The H2 helix in each dsRBD is important for recognition of pri-miRNA substrates. This structure, together with fluorescent resonance energy transfer and mutational analyses, suggests that the DGCR8 core recognizes pri-miRNA in two possible orientations. We propose a model for DGCR8's recognition of pri-miRNA.


Subject(s)
Proteins/chemistry , Crystallography, X-Ray , Electrophoretic Mobility Shift Assay , Fluorescence Resonance Energy Transfer , Humans , MicroRNAs/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Proteins/genetics , Proteins/metabolism , RNA-Binding Proteins
16.
Nucleic Acids Res ; 34(16): 4622-9, 2006.
Article in English | MEDLINE | ID: mdl-16963499

ABSTRACT

DGCR8/Pasha is an essential cofactor for Drosha, a nuclear RNase III that cleaves the local hairpin structures embedded in long primary microRNA transcripts (pri-miRNAs) in eukaryotes. Although our knowledge of pri-miRNA processing has significantly advanced in recent years, the precise role of DGCR8 in this pathway remains unclear. In our present study, we dissect the domains in DGCR8 that contribute to the processing of pri-miRNAs and the subcellular localization of DGCR8. Drosha is stabilized through an interaction between its middle domain and the conserved C-terminal domain of DGCR8. Furthermore, DGCR8, but not Drosha, can directly and stably interact with pri-miRNAs, and the tandem dsRNA-binding domains (dsRBDs) in DGCR8 are responsible for this recognition. Moreover, the DGCR8 N-terminal region upstream of its dsRBDs is unnecessary for pri-miRNA processing but is critical for nuclear localization. Our study thus provides further insights into the mechanism of action of the Drosha-DGCR8 complex in pri-miRNA processing.


Subject(s)
MicroRNAs/metabolism , Proteins/chemistry , RNA Processing, Post-Transcriptional , Ribonuclease III/metabolism , Amino Acid Sequence , Binding Sites , Cell Line , Cell Nucleus/chemistry , Conserved Sequence , Humans , Mutation , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , RNA-Binding Proteins
17.
Cell ; 125(5): 887-901, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16751099

ABSTRACT

The Drosha-DGCR8 complex initiates microRNA maturation by precise cleavage of the stem loops that are embedded in primary transcripts (pri-miRNAs). Here we propose a model for this process that is based upon evidence from both computational and biochemical analyses. A typical metazoan pri-miRNA consists of a stem of approximately 33 bp, with a terminal loop and flanking segments. The terminal loop is unessential, whereas the flanking ssRNA segments are critical for processing. The cleavage site is determined mainly by the distance (approximately 11 bp) from the stem-ssRNA junction. Purified DGCR8, but not Drosha, interacts with pri-miRNAs both directly and specifically, and the flanking ssRNA segments are vital for this binding to occur. Thus, DGCR8 may function as the molecular anchor that measures the distance from the dsRNA-ssRNA junction. Our current study thus facilitates the prediction of novel microRNAs and will assist in the rational design of small hairpin RNAs for RNA interference.


Subject(s)
MicroRNAs/metabolism , Proteins/metabolism , RNA Interference/physiology , RNA Precursors/metabolism , Ribonuclease III/metabolism , Animals , Base Sequence/genetics , Cell Line , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , Models, Molecular , Oligonucleotides/genetics , Proteins/chemistry , Proteins/genetics , RNA Precursors/genetics , RNA Processing, Post-Transcriptional , RNA Stability/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/chemistry , Ribonuclease III/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...