Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 168(6): 1000-1014.e15, 2017 Mar 09.
Article in English | MEDLINE | ID: mdl-28283057

ABSTRACT

Super-enhancers are an emerging subclass of regulatory regions controlling cell identity and disease genes. However, their biological function and impact on miRNA networks are unclear. Here, we report that super-enhancers drive the biogenesis of master miRNAs crucial for cell identity by enhancing both transcription and Drosha/DGCR8-mediated primary miRNA (pri-miRNA) processing. Super-enhancers, together with broad H3K4me3 domains, shape a tissue-specific and evolutionarily conserved atlas of miRNA expression and function. CRISPR/Cas9 genomics revealed that super-enhancer constituents act cooperatively and facilitate Drosha/DGCR8 recruitment and pri-miRNA processing to boost cell-specific miRNA production. The BET-bromodomain inhibitor JQ1 preferentially inhibits super-enhancer-directed cotranscriptional pri-miRNA processing. Furthermore, super-enhancers are characterized by pervasive interaction with DGCR8/Drosha and DGCR8/Drosha-regulated mRNA stability control, suggesting unique RNA regulation at super-enhancers. Finally, super-enhancers mark multiple miRNAs associated with cancer hallmarks. This study presents principles underlying miRNA biology in health and disease and an unrecognized higher-order property of super-enhancers in RNA processing beyond transcription.


Subject(s)
Enhancer Elements, Genetic , MicroRNAs/metabolism , Animals , Azepines/pharmacology , Gene Expression Regulation , Histone Code , Humans , Mice , Neoplasms/genetics , Organ Specificity , RNA Processing, Post-Transcriptional/drug effects , Transcription Factors/metabolism , Transcription, Genetic , Triazoles/pharmacology
2.
Mol Cell ; 83(11): 1810-1826.e8, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267903

ABSTRACT

Microprocessor (MP), DROSHA-DGCR8, processes primary miRNA transcripts (pri-miRNAs) to initiate miRNA biogenesis. The canonical cleavage mechanism of MP has been extensively investigated and comprehensively validated for two decades. However, this canonical mechanism cannot account for the processing of certain pri-miRNAs in animals. In this study, by conducting high-throughput pri-miRNA cleavage assays for approximately 260,000 pri-miRNA sequences, we discovered and comprehensively characterized a noncanonical cleavage mechanism of MP. This noncanonical mechanism does not need several RNA and protein elements essential for the canonical mechanism; instead, it utilizes previously unrecognized DROSHA dsRNA recognition sites (DRESs). Interestingly, the noncanonical mechanism is conserved across animals and plays a particularly significant role in C. elegans. Our established noncanonical mechanism elucidates MP cleavage in numerous RNA substrates unaccounted for by the canonical mechanism in animals. This study suggests a broader substrate repertoire of animal MPs and an expanded regulatory landscape for miRNA biogenesis.


Subject(s)
MicroRNAs , Animals , MicroRNAs/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism , RNA, Double-Stranded , RNA Processing, Post-Transcriptional
3.
Genes Dev ; 37(9-10): 383-397, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37236670

ABSTRACT

DROSHA serves as a gatekeeper of the microRNA (miRNA) pathway by processing primary transcripts (pri-miRNAs). While the functions of structured domains of DROSHA have been well documented, the contribution of N-terminal proline-rich disordered domain (PRD) remains elusive. Here we show that the PRD promotes the processing of miRNA hairpins located within introns. We identified a DROSHA isoform (p140) lacking the PRD, which is produced by proteolytic cleavage. Small RNA sequencing revealed that p140 is significantly impaired in the maturation of intronic miRNAs. Consistently, our minigene constructs demonstrated that PRD enhances the processing of intronic hairpins, but not those in exons. Splice site mutations did not affect the PRD's enhancing effect on intronic constructs, suggesting that the PRD acts independently of splicing reaction by interacting with sequences residing within introns. The N-terminal regions from zebrafish and Xenopus DROSHA can replace the human counterpart, indicating functional conservation despite poor sequence alignment. Moreover, we found that rapidly evolving intronic miRNAs are generally more dependent on PRD than conserved ones, suggesting a role of PRD in miRNA evolution. Our study reveals a new layer of miRNA regulation mediated by a low-complexity disordered domain that senses the genomic contexts of miRNA loci.


Subject(s)
MicroRNAs , Ribonuclease III , Animals , Humans , Introns/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Proline/genetics , Proline/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , RNA Processing, Post-Transcriptional , Zebrafish
4.
Mol Cell ; 81(16): 3422-3439.e11, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34320405

ABSTRACT

Maturation of canonical microRNA (miRNA) is initiated by DROSHA that cleaves the primary transcript (pri-miRNA). More than 1,800 miRNA loci are annotated in humans, but it remains largely unknown whether and at which sites pri-miRNAs are cleaved by DROSHA. Here, we performed in vitro processing on a full set of human pri-miRNAs (miRBase version 21) followed by sequencing. This comprehensive profiling enabled us to classify miRNAs on the basis of DROSHA dependence and map their cleavage sites with respective processing efficiency measures. Only 758 pri-miRNAs are confidently processed by DROSHA, while the majority may be non-canonical or false entries. Analyses of the DROSHA-dependent pri-miRNAs show key cis-elements for processing. We observe widespread alternative processing and unproductive cleavage events such as "nick" or "inverse" processing. SRSF3 is a broad-acting auxiliary factor modulating alternative processing and suppressing unproductive processing. The profiling data and methods developed in this study will allow systematic analyses of miRNA regulation.


Subject(s)
MicroRNAs/genetics , RNA Processing, Post-Transcriptional/genetics , Ribonuclease III/genetics , Serine-Arginine Splicing Factors/genetics , Binding Sites/genetics , Genome, Human/genetics , HEK293 Cells , Humans , RNA Interference
5.
Mol Cell ; 78(2): 303-316.e4, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302542

ABSTRACT

Nuclear processing of most miRNAs is mediated by Microprocessor, comprised of RNase III enzyme Drosha and its cofactor DGCR8. Here, we uncover a hidden layer of Microprocessor regulation via studies of Dicer-independent mir-451, which is clustered with canonical mir-144. Although mir-451 is fully dependent on Drosha/DGCR8, its short stem and small terminal loop render it an intrinsically weak Microprocessor substrate. Thus, it must reside within a cluster for normal biogenesis, although the identity and orientation of its neighbor are flexible. We use DGCR8 tethering assays and operon structure-function assays to demonstrate that local recruitment and transfer of Microprocessor enhances suboptimal substrate processing. This principle applies more broadly since genomic analysis indicates suboptimal canonical miRNAs are enriched in operons, and we validate several of these experimentally. Proximity-based enhancement of suboptimal hairpin processing provides a rationale for genomic retention of certain miRNA operons and may explain preferential evolutionary emergence of miRNA operons.


Subject(s)
Genomics , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Ribonuclease III/genetics , Cell Nucleus/genetics , Humans , RNA Processing, Post-Transcriptional/genetics
6.
Mol Cell ; 78(3): 423-433.e5, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32220645

ABSTRACT

A commencing and critical step in miRNA biogenesis involves processing of pri-miRNAs in the nucleus by Microprocessor. An important, but not completely understood, question is how Drosha, the catalytic subunit of Microprocessor, binds pri-miRNAs and correctly specifies cleavage sites. Here we report the cryoelectron microscopy structures of the Drosha-DGCR8 complex with and without a pri-miRNA. The RNA-bound structure provides direct visualization of the tertiary structure of pri-miRNA and shows that a helix hairpin in the extended PAZ domain and the mobile basic (MB) helix in the RNase IIIa domain of Drosha coordinate to recognize the single-stranded to double-stranded junction of RNA, whereas the dsRNA binding domain makes extensive contacts with the RNA stem. Furthermore, the RNA-free structure reveals an autoinhibitory conformation of the PAZ helix hairpin. These findings provide mechanistic insights into pri-miRNA cleavage site selection and conformational dynamics governing pri-miRNA recognition by the catalytic component of Microprocessor.


Subject(s)
MicroRNAs/chemistry , MicroRNAs/metabolism , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Animals , Cryoelectron Microscopy , Humans , Models, Molecular , Protein Conformation , Protein Domains , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Spodoptera/cytology
7.
Mol Cell ; 78(2): 289-302.e6, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302541

ABSTRACT

Microprocessor initiates the processing of microRNAs (miRNAs) from the hairpin regions of primary transcripts (pri-miRNAs). Pri-miRNAs often contain multiple miRNA hairpins, and this clustered arrangement can assist in the processing of otherwise defective hairpins. We find that miR-451, which derives from a hairpin with a suboptimal terminal loop and a suboptimal stem length, accumulates to 40-fold higher levels when clustered with a helper hairpin. This phenomenon tolerates changes in hairpin order, linker lengths, and the identities of the helper hairpin, the recipient hairpin, the linker-sequence, and the RNA polymerase that transcribes the hairpins. It can act reciprocally and need not occur co-transcriptionally. It requires Microprocessor recognition of the helper hairpin and linkage of the two hairpins, yet predominantly manifests after helper-hairpin processing. It also requires enhancer of rudimentary homolog (ERH), which copurifies with Microprocessor and can dimerize and interact with other proteins that can dimerize, suggesting a model in which one Microprocessor recruits another Microprocessor.


Subject(s)
Cell Cycle Proteins/genetics , MicroRNAs/genetics , RNA Polymerase III/genetics , Transcription Factors/genetics , DNA-Directed RNA Polymerases/genetics , Gene Expression Regulation/genetics , Humans , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription, Genetic
8.
Mol Cell ; 78(3): 411-422.e4, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32220646

ABSTRACT

Metazoan microRNAs require specific maturation steps initiated by Microprocessor, comprising Drosha and DGCR8. Lack of structural information for the assembled complex has hindered an understanding of how Microprocessor recognizes primary microRNA transcripts (pri-miRNAs). Here we present a cryoelectron microscopy structure of human Microprocessor with a pri-miRNA docked in the active site, poised for cleavage. The basal junction is recognized by a four-way intramolecular junction in Drosha, triggered by the Belt and Wedge regions that clamp over the ssRNA. The belt is important for efficiency and accuracy of pri-miRNA processing. Two dsRBDs form a molecular ruler to measure the stem length between the two dsRNA-ssRNA junctions. The specific organization of the dsRBDs near the apical junction is independent of Drosha core domains, as observed in a second structure in the partially docked state. Collectively, we derive a molecular model to explain how Microprocessor recognizes a pri-miRNA and accurately identifies the cleavage site.


Subject(s)
MicroRNAs/chemistry , RNA-Binding Proteins/chemistry , Ribonuclease III/chemistry , Cryoelectron Microscopy , Humans , MicroRNAs/metabolism , Models, Molecular , Protein Conformation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism
9.
Mol Cell ; 80(5): 892-902.e4, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33188727

ABSTRACT

Primary microRNAs (miRNAs) are the precursors of miRNAs that modulate the expression of most mRNAs in humans. They fold up into a hairpin structure that is cleaved at its base by an enzyme complex known as the Microprocessor (Drosha/DGCR8). While many of the molecular details are known, a complete understanding of what features distinguish primary miRNA from hairpin structures in other transcripts is still lacking. We develop a massively parallel functional assay termed Dro-seq (Drosha sequencing) that enables testing of hundreds of known primary miRNA substrates and thousands of single-nucleotide variants. We find an additional feature of primary miRNAs, called Shannon entropy, describing the structural ensemble important for processing. In a deep mutagenesis experiment, we observe particular apical loop U bases, likely recognized by DGCR8, are important for efficient processing. These findings build on existing knowledge about primary miRNA maturation by the Microprocessor and further explore the substrate RNA sequence-structure relationship.


Subject(s)
MicroRNAs , Multiprotein Complexes , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional , RNA-Binding Proteins , Ribonuclease III , Animals , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Sf9 Cells , Spodoptera
10.
Mol Cell ; 78(5): 876-889.e6, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32502422

ABSTRACT

Many microRNAs (miRNAs) are generated from primary transcripts containing multiple clustered stem-loop structures that are thought to be recognized and cleaved by the Microprocessor complex as independent units. Here, we uncover an unexpected mode of processing of the bicistronic miR-15a-16-1 cluster. We find that the primary miR-15a stem-loop is not processed on its own but that the presence of the neighboring primary miR-16-1 stem-loop on the same transcript can compensate for this deficiency in cis. Using a CRISPR/Cas9 screen, we identify SAFB2 (scaffold attachment factor B2) as an essential co-factor in this miR-16-1-assisted pri-miR-15 cleavage and describe SAFB2 as an accessory protein of the Microprocessor. Notably, SAFB2-mediated cleavage expands to other clustered pri-miRNAs, indicating a general mechanism. Together, our study reveals an unrecognized function of SAFB2 in miRNA processing and suggests a scenario in which SAFB2 enables the binding and processing of suboptimal Microprocessor substrates in clustered primary miRNA transcripts.


Subject(s)
Matrix Attachment Region Binding Proteins/metabolism , MicroRNAs/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Receptors, Estrogen/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , HEK293 Cells , Humans , Inverted Repeat Sequences/genetics , Inverted Repeat Sequences/physiology , Matrix Attachment Region Binding Proteins/genetics , Mice , MicroRNAs/genetics , Nuclear Matrix-Associated Proteins/genetics , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/metabolism , Receptors, Estrogen/genetics
11.
Mol Cell ; 73(3): 505-518.e5, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30554947

ABSTRACT

Microprocessor, composed of DROSHA and its cofactor DGCR8, initiates microRNA (miRNA) biogenesis by processing the primary transcripts of miRNA (pri-miRNAs). Here we investigate the mechanism by which Microprocessor selects the cleavage site with single-nucleotide precision, which is crucial for the specificity and functionality of miRNAs. By testing ∼40,000 pri-miRNA variants, we find that for some pri-miRNAs the cleavage site is dictated mainly by the mGHG motif embedded in the lower stem region of pri-miRNA. Structural modeling and deep-sequencing-based complementation experiments show that the double-stranded RNA-binding domain (dsRBD) of DROSHA recognizes mGHG to place the catalytic center in the appropriate position. The mGHG motif as well as the mGHG-recognizing residues in DROSHA dsRBD are conserved across eumetazoans, suggesting that this mechanism emerged in an early ancestor of the animal lineage. Our findings provide a basis for the understanding of miRNA biogenesis and rational design of accurate small-RNA-based gene silencing.


Subject(s)
MicroRNAs/metabolism , Nucleotide Motifs , RNA Processing, Post-Transcriptional , Ribonuclease III/metabolism , HCT116 Cells , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Interaction Domains and Motifs , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Structure-Activity Relationship , Substrate Specificity
12.
Mol Cell ; 75(2): 340-356.e10, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31253575

ABSTRACT

The microRNAs encoded by the miR-17∼92 polycistron are commonly overexpressed in cancer and orchestrate a wide range of oncogenic functions. Here, we identify a mechanism for miR-17∼92 oncogenic function through the disruption of endogenous microRNA (miRNA) processing. We show that, upon oncogenic overexpression of the miR-17∼92 primary transcript (pri-miR-17∼92), the microprocessor complex remains associated with partially processed intermediates that aberrantly accumulate. These intermediates reflect a series of hierarchical and conserved steps in the early processing of the pri-miR-17∼92 transcript. Encumbrance of the microprocessor by miR-17∼92 intermediates leads to the broad but selective downregulation of co-expressed polycistronic miRNAs, including miRNAs derived from tumor-suppressive miR-34b/c and from the Dlk1-Dio3 polycistrons. We propose that the identified steps of polycistronic miR-17∼92 biogenesis contribute to the oncogenic re-wiring of gene regulation networks. Our results reveal previously unappreciated functional paradigms for polycistronic miRNAs in cancer.


Subject(s)
Carcinogenesis/genetics , MicroRNAs/genetics , RNA Processing, Post-Transcriptional/genetics , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Iodide Peroxidase/genetics , Membrane Proteins/genetics , MicroRNAs/biosynthesis , Nucleic Acid Conformation
13.
Proc Natl Acad Sci U S A ; 120(41): e2306727120, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37788316

ABSTRACT

The nuclear cleavage of a suboptimal primary miRNA hairpin by the Drosha/DGCR8 complex ("Microprocessor") can be enhanced by an optimal miRNA neighbor, a phenomenon termed cluster assistance. Several features and biological impacts of this new layer of miRNA regulation are not fully known. Here, we elucidate the parameters of cluster assistance of a suboptimal miRNA and also reveal competitive interactions amongst optimal miRNAs within a cluster. We exploit cluster assistance as a functional assay for suboptimal processing and use this to invalidate putative suboptimal substrates, as well as identify a "solo" suboptimal miRNA. Finally, we report complexity in how specific mutations might affect the biogenesis of clustered miRNAs in disease contexts. This includes how an operon context can buffer the effect of a deleterious processing variant, but reciprocally how a point mutation can have a nonautonomous effect to impair the biogenesis of a clustered, suboptimal, neighbor. These data expand our knowledge regarding regulated miRNA biogenesis in humans and represent a functional assay for empirical definition of suboptimal Microprocessor substrates.


Subject(s)
MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism
14.
RNA ; 29(8): 1166-1184, 2023 08.
Article in English | MEDLINE | ID: mdl-37169394

ABSTRACT

The potential for microRNAs (miRNAs) to regulate gene expression remains incompletely understood. DROSHA initiates the biogenesis of miRNAs while variants of Argonaute (AGO) and trinucleotide repeat containing six (TNRC6) family proteins form complexes with miRNAs to facilitate RNA recognition and gene regulation. Here we investigate the fate of miRNAs in the absence of these critical RNAi protein factors. Knockout of DROSHA expression reduces levels of some miRNAs annotated in miRBase but not others. The identity of miRNAs with reduced expression matches the identity of miRNAs previously identified by experimental approaches. The MirGeneDB resource offers the closest alignment with experimental results. In contrast, the loss of TNRC6 proteins had much smaller effects on miRNA levels. Knocking out AGO proteins, which directly contact the mature miRNA, decreased expression of the miRNAs most strongly associated with AGO2 as determined from enhanced crosslinking immunoprecipitation (AGO2-eCLIP). Evaluation of miRNA binding to endogenously expressed AGO proteins revealed that miRNA:AGO association was similar for AGO1, AGO2, AGO3, and AGO4. Our data emphasize the need to evaluate annotated miRNAs based on approximate cellular abundance, DROSHA-dependence, and physical association with AGO when forming hypotheses related to their function.


Subject(s)
MicroRNAs , MicroRNAs/metabolism , RNA Interference , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Gene Expression Regulation , Trinucleotide Repeats
15.
Mol Cell ; 66(2): 258-269.e5, 2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28431232

ABSTRACT

MicroRNA (miRNA) maturation is initiated by DROSHA, a double-stranded RNA (dsRNA)-specific RNase III enzyme. By cleaving primary miRNAs (pri-miRNAs) at specific positions, DROSHA serves as a main determinant of miRNA sequences and a highly selective gatekeeper for the canonical miRNA pathway. However, the sites of DROSHA-mediated processing have not been annotated, and it remains unclear to what extent DROSHA functions outside the miRNA pathway. Here, we establish a protocol termed "formaldehyde crosslinking, immunoprecipitation, and sequencing (fCLIP-seq)," which allows identification of DROSHA cleavage sites at single-nucleotide resolution. fCLIP identifies numerous processing sites, suggesting widespread end modifications during miRNA maturation. fCLIP also finds many pri-miRNAs that undergo alternative processing, yielding multiple miRNA isoforms. Moreover, we discovered dozens of DROSHA substrates on non-miRNA loci, which may serve as cis-elements for DROSHA-mediated gene regulation. We anticipate that fCLIP-seq could be a general tool for investigating interactions between dsRNA-binding proteins and structured RNAs.


Subject(s)
High-Throughput Nucleotide Sequencing , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Ribonuclease III/metabolism , Sequence Analysis, RNA/methods , Base Sequence , Binding Sites , Cross-Linking Reagents/chemistry , Formaldehyde/chemistry , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , MicroRNAs/chemistry , MicroRNAs/genetics , Nucleic Acid Conformation , Protein Binding , RNA Interference , Ribonuclease III/chemistry , Ribonuclease III/genetics , Structure-Activity Relationship , Substrate Specificity , Transfection
16.
Liver Int ; 44(7): 1634-1650, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38517158

ABSTRACT

BACKGROUND: Excessive fatty acids in the liver lead to the accumulation of lipotoxic lipids and then cellular stress to further evoke the related disease, like non-alcoholic fatty liver disease (NAFLD). As reported, fatty acid stimulation can cause some specific miRNA dysregulation, which caused us to investigate the relationship between miRNA biogenesis and fatty acid overload. METHODS: Gene expression omnibus (GEO) dataset analysis, miRNA-seq, miRNA cleavage assay, RT-qPCR, western blotting, immunofluorescence and co-immunoprecipitation (co-IP) were used to reveal the change of miRNAs under pathological status and explore the relevant mechanism. High fat, high fructose, high cholesterol (HFHFrHC) diet-fed mice transfected with AAV2/8-shDrosha or AAV2/8-shPRMT5 were established to investigate the in vivo effects of Drosha or PRMT5 on NAFLD phenotype. RESULTS: We discovered that the cleavage of miRNAs was inhibited by analysing miRNA contents and detecting some representative pri-miRNAs in multiple mouse and cell models, which was further verified by the reduction of the Microprocessor activity in the presence of palmitic acid (PA). In vitro, PA could induce Drosha, the core RNase III in the Microprocessor complex, degrading through the proteasome-mediated pathway, while in vivo, knockdown of Drosha significantly promoted NAFLD to develop to a more serious stage. Mechanistically, our results demonstrated that PA can increase the methyltransferase activity of PRMT5 to degrade Drosha through MDM2, a ubiquitin E3 ligase for Drosha. The above results indicated that PRMT5 may be a critical regulator in lipid metabolism during NAFLD, which was confirmed by the knocking down of PRMT5 improved aberrant lipid metabolism in vitro and in vivo. CONCLUSIONS: We first demonstrated the relationship between miRNA dosage and NAFLD and proved that PA can activate the PRMT5-MDM2-Drosha signalling pathway to regulate miRNA biogenesis.


Subject(s)
Lipid Metabolism , MicroRNAs , Non-alcoholic Fatty Liver Disease , Protein-Arginine N-Methyltransferases , Proto-Oncogene Proteins c-mdm2 , Animals , Humans , Male , Mice , Diet, High-Fat , Disease Models, Animal , Fatty Acids/metabolism , Liver/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Ribonuclease III/metabolism , Ribonuclease III/genetics , Signal Transduction
17.
Mol Cell ; 63(3): 420-32, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27425409

ABSTRACT

Recent studies suggest that the microprocessor (Drosha-DGCR8) complex can be recruited to chromatin to catalyze co-transcriptional processing of primary microRNAs (pri-miRNAs) in mammalian cells. However, the molecular mechanism of co-transcriptional miRNA processing is poorly understood. Here we find that HP1BP3, a histone H1-like chromatin protein, specifically associates with the microprocessor and promotes global miRNA biogenesis in human cells. Chromatin immunoprecipitation (ChIP) studies reveal genome-wide co-localization of HP1BP3 and Drosha and HP1BP3-dependent Drosha binding to actively transcribed miRNA loci. Moreover, HP1BP3 specifically binds endogenous pri-miRNAs and facilitates the Drosha/pri-miRNA association in vivo. Knockdown of HP1BP3 compromises pri-miRNA processing by causing premature release of pri-miRNAs from the chromatin. Taken together, these studies suggest that HP1BP3 promotes co-transcriptional miRNA processing via chromatin retention of nascent pri-miRNA transcripts. This work significantly expands the functional repertoire of the H1 family of proteins and suggests the existence of chromatin retention factors for widespread co-transcriptional miRNA processing.


Subject(s)
Chromatin/metabolism , MicroRNAs/biosynthesis , Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , Transcription, Genetic , Animals , Binding Sites , Chromatin/genetics , Chromatin Immunoprecipitation , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA-Binding Proteins , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Genome, Human , HeLa Cells , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics , Protein Binding , RNA Interference , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Transfection
18.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062888

ABSTRACT

Mirtrons represent a subclass of microRNAs (miRNAs) that rely on the splicing machinery for their maturation. However, the molecular details of this Drosha-independent processing are still not fully understood; as an example, the Microprocessor complex cannot process the mirtronic pre-miRNA from the transcript even if splice site mutations are present. To investigate the influence of alternative splicing sites on mirtron formation, we generated Enhanced Green Fluorescent Protein (EGFP) reporters containing artificial introns to compare the processing of canonical miRNAs and mirtrons. Although mutations of both splice sites generated a complex pattern of alternative transcripts, mirtron formation was always severely affected as opposed to the normal processing of the canonical hsa-mir-33b miRNA. However, we also detected that while its formation was also hindered, the mirtron-derived hsa-mir-877-3p miRNA was less affected by certain mutations than the hsa-mir-877-5p species. By knocking down Drosha, we showed that this phenomenon is not dependent on Microprocessor activity but rather points toward the potential stability difference between the miRNAs from the different arms. Our results indicate that when the major splice sites are mutated, mirtron formation cannot be rescued by nearby alternative splice sites, and stability differences between 5p and 3p species should also be considered for functional studies of mirtrons.


Subject(s)
Alternative Splicing , MicroRNAs , Ribonuclease III , MicroRNAs/genetics , Humans , Ribonuclease III/genetics , Ribonuclease III/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splice Sites/genetics , Mutation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Introns/genetics
19.
J Cell Sci ; 134(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34137440

ABSTRACT

Hypoxia is a severe stressor to cellular homeostasis. At the cellular level, low oxygen triggers the transcription of a variety of genes supporting cell survival and oxygen homeostasis mediated by transcription factors, such as hypoxia-inducible factors (HIFs). Among many determinants dictating cell responses to hypoxia and HIFs are microRNAs (miRNAs). Cajal bodies (CBs), subnuclear structures involved in ribonucleoprotein biogenesis, have been recently proven to contribute to miRNA processing and biogenesis but have not been studied under hypoxia. Here, we show, for the first time, a hypoxia-dependent increase in CB number in WI-38 primary fibroblasts, which normally have very few CBs. Additionally, the CB marker protein coilin is upregulated in hypoxic WI-38 cells. However, the hypoxic coilin upregulation was not seen in transformed cell lines. Furthermore, we found that coilin is needed for the hypoxic induction of a well-known hypoxia-induced miRNA (hypoxamiR), miR-210, as well as for the hypoxia-induced alternative splicing of the miR-210 host gene, MIR210HG. These findings provide a new link in the physiological understanding of coilin, CBs and miRNA dysregulation in hypoxic pathology.


Subject(s)
MicroRNAs , Alternative Splicing/genetics , Cell Hypoxia , Coiled Bodies/genetics , Coiled Bodies/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Transcription Factors/metabolism
20.
J Cell Sci ; 134(6)2021 03 22.
Article in English | MEDLINE | ID: mdl-33558311

ABSTRACT

The DNA damage response (DDR) is the signaling cascade that recognizes DNA double-strand breaks (DSBs) and promotes their resolution via the DNA repair pathways of non-homologous end joining (NHEJ) or homologous recombination (HR). We and others have shown that DDR activation requires DROSHA; however, whether DROSHA exerts its functions by associating with damage sites, what controls its recruitment, and how DROSHA influences DNA repair remains poorly understood. Here, we show that DROSHA associates with DSBs independently of transcription. Neither H2AX, nor ATM or DNA-PK kinase activities are required for recruitment of DROSHA to break sites. Rather, DROSHA interacts with RAD50, and inhibition of the MRN complex by mirin treatment abolishes this interaction. MRN complex inactivation by RAD50 knockdown or mirin treatment prevents DROSHA recruitment to DSBs and, as a consequence, also prevents 53BP1 (also known as TP53BP1) recruitment. During DNA repair, DROSHA inactivation reduces NHEJ and boosts HR frequency. Indeed, DROSHA knockdown also increases the association of downstream HR factors such as RAD51 to DNA ends. Overall, our results demonstrate that DROSHA is recruited at DSBs by the MRN complex and directs DNA repair towards NHEJ.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Damage/genetics , DNA Repair/genetics , Homologous Recombination
SELECTION OF CITATIONS
SEARCH DETAIL