Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
Int J Mol Sci ; 22(18)2021 Sep 12.
Article in English | MEDLINE | ID: mdl-34576018

ABSTRACT

RNAi-mediated knockdown of DICER1 and DROSHA, enzymes critically involved in miRNA biogenesis, has been postulated to affect the homeostasis and the angiogenic capacity of human endothelial cells. To re-evaluate this issue, we reduced the expression of DICER1 or DROSHA by RNAi-mediated knockdown and subsequently investigated the effect of these interventions on the angiogenic capacity of human umbilical vein endothelial cells (HUVEC) in vitro (proliferation, migration, tube formation, endothelial cell spheroid sprouting) and in a HUVEC xenograft assay in immune incompetent NSGTM mice in vivo. In contrast to previous reports, neither knockdown of DICER1 nor knockdown of DROSHA profoundly affected migration or tube formation of HUVEC or the angiogenic capacity of HUVEC in vivo. Furthermore, knockdown of DICER1 and the combined knockdown of DICER1 and DROSHA tended to increase VEGF-induced BrdU incorporation and induced angiogenic sprouting from HUVEC spheroids. Consistent with these observations, global proteomic analyses showed that knockdown of DICER1 or DROSHA only moderately altered HUVEC protein expression profiles but additively reduced, for example, expression of the angiogenesis inhibitor thrombospondin-1. In conclusion, global reduction of miRNA biogenesis by knockdown of DICER1 or DROSHA does not inhibit the angiogenic capacity of HUVEC. Further studies are therefore needed to elucidate the influence of these enzymes in the context of human endothelial cell-related angiogenesis.


Subject(s)
DEAD-box RNA Helicases/physiology , Endothelial Cells/physiology , Neovascularization, Physiologic , Ribonuclease III/physiology , Animals , Humans
2.
Acta Biochim Biophys Sin (Shanghai) ; 53(4): 463-471, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33751023

ABSTRACT

A large number of proteins involved in RNA metabolism possess a double-stranded RNA-binding domain (dsRBD), whose sequence variations and functional versatilities are still being recognized. All dsRBDs have a similar structural fold: α1-L1-ß1-L2-ß2-L3-ß3-L4-α2 (α represents an α-helix, ß a ß-sheet, and L a loop conformation between the well-defined secondary structures). Our recent work revealed that the dsRBD in Drosha, which is involved in animal microRNA (miRNA) biogenesis, differs from other dsRBDs by containing a short insertion in its L1 region and that this insertion is important for Drosha function. We asked why the same insertion is excluded in all other dsRBDs and proposed that a longer L1 may be detrimental to their functions. In this study, to test this hypothesis, we inserted the Drosha sequence into several well-known dsRBDs from various organisms. Gel mobility shift assay demonstrated that L1 extension invariably reduced RNA binding by these dsRBDs. In addition, such a mutation in Dicer, another protein involved in miRNA biogenesis, impaired Dicer's ability to process miRNAs, which led to de-repression of reporter expression, in human cells. Taken together, our results add to the growing appreciation of the diversity in dsRBDs and suggest that dsRBDs have intricate structures and functions that are sensitive to perturbations in the L1 region.


Subject(s)
Double-Stranded RNA Binding Motif , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Amino Acid Sequence , Animals , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/physiology , DNA, Single-Stranded/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/physiology , HEK293 Cells , Humans , MicroRNAs/metabolism , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/physiology , Protein Structure, Secondary , RNA/metabolism , RNA-Binding Proteins/genetics , Ribonuclease III/chemistry , Ribonuclease III/genetics , Ribonuclease III/physiology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus Proteins/physiology
3.
Nucleic Acids Res ; 48(13): 7135-7153, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32484548

ABSTRACT

Constitutive heterochromatin at the pericentric regions of chromosomes undergoes dynamic changes in its epigenetic and spatial organization during spermatogenesis. Accurate control of pericentric heterochromatin is required for meiotic cell divisions and production of fertile and epigenetically intact spermatozoa. In this study, we demonstrate that pericentric heterochromatin is expressed during mouse spermatogenesis to produce major satellite repeat (MSR) transcripts. We show that the endonuclease DICER localizes to the pericentric heterochromatin in the testis. Furthermore, DICER forms complexes with MSR transcripts, and their processing into small RNAs is compromised in Dicer1 knockout mice leading to an elevated level of MSR transcripts in meiotic cells. We also show that defective MSR forward transcript processing in Dicer1 cKO germ cells is accompanied with reduced recruitment of SUV39H2 and H3K9me3 to the pericentric heterochromatin and meiotic chromosome missegregation. Altogether, our results indicate that the physiological role of DICER in maintenance of male fertility extends to the regulation of pericentric heterochromatin through direct targeting of MSR transcripts.


Subject(s)
DEAD-box RNA Helicases/physiology , Ribonuclease III/physiology , Spermatids , Spermatocytes , Spermatogenesis , Testis/metabolism , Animals , Centromere/metabolism , Chromosome Segregation , Fertility , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Male , Meiosis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatids/cytology , Spermatids/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism , Tandem Repeat Sequences/genetics , Testis/cytology
4.
Cell Biol Int ; 44(6): 1394-1404, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32129540

ABSTRACT

Hypoxia is a condition in which the whole body or a region of the body is deprived of oxygen supply. The brain is very sensitive to the lack of oxygen and cerebral hypoxia can rapidly cause severe brain damage. Astrocytes are essential for the survival and function of neurons. Therefore, protecting astrocytes against cell death is one of the main therapeutic strategies for treating hypoxia. Hence, the mechanism of hypoxia-induced astrocytic cell death should be fully elucidated. In this study, astrocytes were exposed to hypoxic conditions using a hypoxia work station or the hypoxia mimetic agent cobalt chloride (CoCl2 ). Both the hypoxic gas mixture (1% O2 ) and chemical hypoxia-induced apoptotic cell death in T98G glioblastoma cells and mouse primary astrocytes. Reactive oxygen species were generated in response to the hypoxia-mediated activation of caspase-1. Active caspase-1 induced the classical caspase-dependent apoptosis of astrocytes. In addition, the microRNA processing enzyme Dicer was cleaved by caspase-3 during hypoxia. Knockdown of Dicer using antisense oligonucleotides induced apoptosis of T98G cells. Taken together, these results suggest that astrocytic cell death during hypoxia is mediated by the reactive oxygen species/caspase-1/classical caspase-dependent apoptotic pathway. In addition, the decrease in Dicer levels by active caspase-3 amplifies this apoptotic pathway via a positive feedback loop. These findings may provide a new target for therapeutic interventions in cerebral hypoxia.


Subject(s)
Astrocytes/metabolism , Brain , Caspase 1/metabolism , DEAD-box RNA Helicases/physiology , Ribonuclease III/physiology , Animals , Apoptosis , Astrocytes/cytology , Brain/cytology , Brain/metabolism , Cell Hypoxia , Cells, Cultured , Female , Humans , Mice , Mice, Inbred BALB C , Oxidative Stress , Reactive Oxygen Species/metabolism
5.
Mol Brain ; 13(1): 3, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924235

ABSTRACT

The cellular consequences of N-Methyl-D-Aspartate receptor (NMDAR) stimulation depend on the receptors' subcellular localization. Synaptic NMDARs promote plasticity and survival whereas extrasynaptic NMDARs mediate excitotoxicity and contribute to cell death in neurodegenerative diseases. The mechanisms that couple activation of extrasynaptic NMDARs to cell death remain incompletely understood. We here show that activation of extrasynaptic NMDARs by bath application of NMDA or L-glutamate leads to the upregulation of a group of 19 microRNAs in cultured mouse hippocampal neurons. In contrast, none of these microRNAs is induced upon stimulation of synaptic activity. Increased microRNA expression depends on the pri-miRNA processing enzyme Drosha, but not on de novo gene transcription. These findings suggest that toxic NMDAR signaling involves changes in the expression levels of particular microRNAs.


Subject(s)
MicroRNAs/genetics , N-Methylaspartate/toxicity , Neurotoxins/toxicity , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/genetics , Transcriptome , Animals , Bicuculline/pharmacology , Cells, Cultured , GABA-A Receptor Antagonists/pharmacology , Glutamic Acid/pharmacology , Glycine/pharmacology , Glycine/toxicity , Hippocampus/cytology , Kainic Acid/toxicity , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , N-Methylaspartate/pharmacology , Neurotoxins/pharmacology , Rats, Sprague-Dawley , Ribonuclease III/physiology , Seizures/chemically induced , Specific Pathogen-Free Organisms , Subcellular Fractions/metabolism , Up-Regulation/drug effects
6.
Cells ; 8(8)2019 08 20.
Article in English | MEDLINE | ID: mdl-31434287

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) has been linked to chronic lung inflammation. Drosha ribonuclease III (DROSHA), a class 2 ribonuclease III enzyme, plays a key role in microRNA (miRNA) biogenesis. However, the mechanisms by which DROSHA affects the lung inflammation during idiopathic pulmonary fibrosis (IPF) remain unclear. Here, we demonstrate that DROSHA regulates the absent in melanoma 2 (AIM2) inflammasome activation during idiopathic pulmonary fibrosis (IPF). Both DROSHA and AIM2 protein expression were elevated in alveolar macrophages of patients with IPF. We also found that DROSHA and AIM2 protein expression were increased in alveolar macrophages of lung tissues in a mouse model of bleomycin-induced pulmonary fibrosis. DROSHA deficiency suppressed AIM2 inflammasome-dependent caspase-1 activation and interleukin (IL)-1ß and IL-18 secretion in primary mouse alveolar macrophages and bone marrow-derived macrophages (BMDMs). Transduction of microRNA (miRNA) increased the formation of the adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) specks, which is required for AIM2 inflammasome activation in BMDMs. Our results suggest that DROSHA promotes AIM2 inflammasome activation-dependent lung inflammation during IPF.


Subject(s)
DNA-Binding Proteins/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Inflammasomes/metabolism , Macrophages, Alveolar/metabolism , Macrophages/metabolism , Pneumonia , Ribonuclease III/physiology , Adult , Aged , Animals , Cells, Cultured , Chronic Disease , Female , Humans , Macrophages/pathology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pneumonia/metabolism , Pneumonia/pathology
7.
Life Sci ; 232: 116656, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31306658

ABSTRACT

AIMS: Tamoxifen-induced liver-specific Dicer1 deletion (iDicer1-/-) in mature mice may provide clues demonstrating the genuine effects of acute loss of Dicer1 and miRNAs in the liver regeneration process. MAIN METHODS: In this study, mice with tamoxifen-induced Dicer1 deletion through the Cre/LoxP system were constructed and then underwent classic 70% partial hepatectomy or CCl4-induced liver injury. To rescue the inhibitory effect of Dicer1 ablation on liver regeneration, miR-21 agomir was injected into the tail vein of iDicer1-/- mice. KEY FINDINGS: Unlike constitutive embryonic deletion of Dicer1, tamoxifen-induced Dicer1 deletion did not result in severe liver injury or lesions, providing an ideal model for investigating acute loss of Dicer1 and miRNAs in liver regeneration. Dicer1 deletion led to impaired liver regeneration through the inhibitory effect of miR-21 on PTEN and Rhob expression. SIGNIFICANCE: In our previous study, we found that embryonic loss of Dicer1 impairs hepatocyte survival and leads to chronic inflammation and progenitor cell activation, while the role of Dicer1 in liver regeneration remains largely unknown. We clearly identified the promotion effect of Dicer1 on liver regeneration by increasing miR-21 expression, which inhibits the expression of two negative cell proliferation regulators, Pten and Rhob.


Subject(s)
DEAD-box RNA Helicases/physiology , Liver Regeneration/physiology , MicroRNAs/physiology , PTEN Phosphohydrolase/metabolism , Ribonuclease III/physiology , rhoB GTP-Binding Protein/metabolism , Animals , DEAD-box RNA Helicases/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Mice, Knockout , Ribonuclease III/genetics , Tamoxifen/administration & dosage
8.
Commun Biol ; 2: 211, 2019.
Article in English | MEDLINE | ID: mdl-31240249

ABSTRACT

Cell cycle progression depends on phase-specific gene expression. Here we show that the nuclear RNA degradation machinery plays a lead role in promoting cell cycle-dependent gene expression by triggering promoter-dependent co-transcriptional RNA degradation. Single molecule quantification of RNA abundance in different phases of the cell cycle indicates that relative curtailment of gene expression in certain phases is attained even when transcription is not completely inhibited. When nuclear ribonucleases are deleted, transcription of the Saccharomyces cerevisiae G1-specific axial budding gene AXL2 is detected throughout the cell cycle and its phase-specific expression is lost. Promoter replacement abolished cell cycle-dependent RNA degradation and rendered the RNA insensitive to the deletion of nuclear ribonucleases. Together the data reveal a model of gene regulation whereby RNA abundance is controlled by promoter-dependent induction of RNA degradation.


Subject(s)
Cell Cycle , Promoter Regions, Genetic , RNA, Nuclear/metabolism , Gene Expression Regulation, Fungal , Humans , Membrane Glycoproteins/genetics , RNA, Messenger/metabolism , Ribonuclease III/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Transcription Factor TFIIA/genetics
9.
Neuromolecular Med ; 21(2): 97-109, 2019 06.
Article in English | MEDLINE | ID: mdl-30963386

ABSTRACT

Studies from last two decades have established microRNAs (miRNAs) as the most influential regulator of gene expression, especially at the post-transcriptional stage. The family of small RNA molecules including miRNAs is highly conserved and expressed throughout the multicellular organism. MiRNAs regulate gene expression by binding to 3' UTR of protein-coding mRNAs and initiating either decay or movement of mRNAs to stress granules. Tissues or cells, which go through cell fate transformation like stem cells, brain cells, iPSCs, or cancer cells show very dynamic expression profile of miRNAs. Inability to pass the developmental stages of Dicer (miRNA maturation enzyme) knockout animals has confirmed that expression of mature and functional miRNAs is essential for proper development of different organs and tissues. Studies from our laboratory and elsewhere have demonstrated the role of miR-200 and miR-34 families in neural development and have shown higher expression of both families in mature and differentiated neurons. In present review, we have provided a general overview of miRNAs and focused on the role of miR-34 and miR-200, two miRNA families, which have the capability to change the phenotype and fate of a cell in different tissues and situations.


Subject(s)
MicroRNAs/genetics , Neurogenesis/genetics , Neuronal Plasticity/physiology , 3' Untranslated Regions , Animals , Cell Differentiation/genetics , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/physiology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Induced Pluripotent Stem Cells/cytology , Mammals , Mice , PC12 Cells , RNA, Untranslated/classification , RNA, Untranslated/genetics , Rats , Ribonuclease III/deficiency , Ribonuclease III/physiology , Terminology as Topic
10.
Biochim Biophys Acta Gene Regul Mech ; 1862(4): 472-485, 2019 04.
Article in English | MEDLINE | ID: mdl-30840854

ABSTRACT

The multidomain RNase III endoribonuclease DICER is required for the generation of most functional microRNAs (miRNAs). Loss of Dicer affects developmental processes at different levels. Here, we characterized the zebrafish Dicer1 mutant, dicer1sa9205, which has a single point mutation induced by N-ethyl-N-nitrosourea mutagenesis. Heterozygous dicer1sa9205 developed normally, being phenotypically indistinguishable from wild-type siblings. Homozygous dicer1sa9205 mutants display smaller eyes, abnormal craniofacial development and aberrant pigmentation. Reduced numbers of both iridophores and melanocytes were observed in the head and ventral trunk of dicer1sa9205 homozygotes; the effect on melanocytes was stronger and detectable earlier in development. The expression of microphthalmia-associated transcription factor a (mitfa), the master gene for melanocytes differentiation, was enhanced in dicer1-depleted fish. Similarly, the expression of SRY-box containing gene 10 (sox10), required for mitfa activation, was higher in mutants than in wild types. In silico and in vivo analyses of either sox10 or mitfa 3'UTRs revealed conserved potential miRNA binding sites likely involved in the post-transcriptional regulation of both genes. Based on these findings, we propose that dicer1 participates in the gene regulatory network governing zebrafish melanocyte differentiation by controlling the expression of mitfa and sox10.


Subject(s)
Cartilage/abnormalities , Melanocytes/cytology , Ribonuclease III/physiology , Zebrafish Proteins/physiology , 3' Untranslated Regions , Animals , Apoptosis , Cartilage/growth & development , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/anatomy & histology , Gene Expression Regulation , Head , Larva/anatomy & histology , Melanocytes/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/metabolism , Mutation , Neural Crest/cytology , Ribonuclease III/genetics , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
Addict Biol ; 24(3): 498-508, 2019 05.
Article in English | MEDLINE | ID: mdl-29516602

ABSTRACT

microRNA (miRNA) play important roles in drug addiction and act as a post-transcriptional regulator of gene expression. We previously reported extensive downregulation of miRNAs in the nucleus accumbens (NAc) of methamphetamine (METH)-sensitized mice. However, the regulatory mechanism of this METH-induced downregulation of miRNAs has yet to be elucidated. Thus, we examined METH-induced changes in the expression of miRNAs and their precursors, as well as the expression levels of mRNA and the proteins involved in miRNA biogenesis such as Dicer1 and Ago2, in the nucleus accumbens of METH-induced locomotor sensitized mice. miRNAs and Ago2 were significantly downregulated, while the expression of miRNA precursors remained unchanged or upregulated, which suggests that the downregulation of miRNAs was likely due to a reduction in Ago2-mediated splicing but unlikely to be regulated at the transcription level. Interestingly, the expression level of Dicer1, which is a potential target of METH-induced decreased miRNAs, such as miR-124, miR-212 and miR-29b, was significantly increased. In conclusion, this study indicates that miRNA biogenesis (such as Ago2 and Dicer1) and their miRNA products may have a role in the development of METH addiction.


Subject(s)
Argonaute Proteins/physiology , Central Nervous System Stimulants/pharmacology , DEAD-box RNA Helicases/physiology , Locomotion/drug effects , Methamphetamine/pharmacology , MicroRNAs/metabolism , Ribonuclease III/physiology , Amphetamine-Related Disorders/physiopathology , Animals , Down-Regulation/drug effects , Male , Mice, Inbred C57BL , Nucleus Accumbens/drug effects
12.
Methods ; 152: 3-11, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29902563

ABSTRACT

CLIP-seq (crosslinking immunoprecipitation and sequencing) is widely used to map the binding sites of a protein of interest on the transcriptome, and generally employs UV to induce the covalent bonds between protein and RNA, which allows stringent washing. However, dsRNA is inefficiently crosslinked by UV, making it difficult to study the interactions between dsRNA binding proteins and their substrates. A dsRNA endoribonuclease DROSHA initiates the maturation of microRNA (miRNA) by cleaving primary miRNA (pri-miRNA). Despite the importance of DROSHA in miRNA maturation and sequence determination, accurate mapping of DROSHA cleavage sites has not been feasible due to rapid processing, modification, and degradation of the cleaved products in cells. Here, we present a high-throughput sequencing method that allows the mapping of in vivo DROSHA cleavage sites at single nucleotide resolution, termed formaldehyde crosslinking, immunoprecipitation, and sequencing (fCLIP-seq). The fCLIP-seq protocol has been improved significantly over the standard CLIP-seq methods by (1) using formaldehyde for efficient and reversible crosslinking, (2) employing polyethylene glycol and adaptors with randomized sequences to enhance ligation efficiency and minimize bias, and (3) performing ligation after elution, which exposes the RNA termini for efficient ligation. fCLIP-seq successfully captures the nascent products of DROSHA, which allows precise mapping of the DROSHA processing sites. Moreover, from the analysis of the distinctive cleavage pattern, we discover previously unknown substrates of DROSHA. fCLIP-seq is a useful tool to obtain transcriptome-wide information on DROSHA activity and can be applied further to investigate other dsRNA-protein interactions.


Subject(s)
Ribonuclease III/chemistry , Sequence Analysis, RNA/methods , HEK293 Cells , HeLa Cells , Humans , RNA-Binding Proteins/chemistry , Ribonuclease III/physiology
13.
Insect Mol Biol ; 28(1): 74-85, 2019 02.
Article in English | MEDLINE | ID: mdl-30171639

ABSTRACT

Nosema ceranae is a microsporidian parasite that infects the honeybee midgut epithelium. The protein-coding gene Dicer is lost in most microsporidian genomes but is present in N. ceranae. By feeding infected honeybees with small interfering RNA targeting the N. ceranae gene coding Dicer (siRNA-Dicer), we found that N. ceranae spore loads were significantly reduced. In addition, over 10% of total parasite protein-coding genes showed significantly divergent expression profiles after siRNA-Dicer treatment. Parasite genes for cell proliferation, ABC transporters and hexokinase were downregulated at 3 days postinfection, a key point in the middle of parasite replication cycles. In addition, genes involved in metabolic pathways of honeybees and N. ceranae showed significant co-expression. Furthermore, the siRNA-Dicer treatment partly reversed the expression patterns of honeybee genes. The honeybee gene mucin-2-like showed significantly upregulation in the siRNA-Dicer group compared with the infection group continually at 4, 5 and 6 days postinfection, suggesting that the siRNA-Dicer feeding promoted the strength of the mucus barrier resulted from interrupted parasite proliferation. As the gene Dicer broadly regulates N. ceranae proliferation and honeybee metabolism, our data suggest the RNA interference pathway is an important infection strategy for N. ceranae.


Subject(s)
Bees/microbiology , Host-Parasite Interactions , Nosema/physiology , Ribonuclease III/physiology , Spores, Fungal , Animals , Bees/genetics , Bees/metabolism , Gene Expression Profiling , Transcriptome
14.
Genes Dev ; 32(7-8): 537-554, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29654060

ABSTRACT

The identification of telomerase-negative HAATI (heterochromatin amplification-mediated and telomerase-independent) cells, in which telomeres are superseded by nontelomeric heterochromatin tracts, challenged the idea that canonical telomeres are essential for chromosome linearity and raised crucial questions as to how such tracts translocate to eroding chromosome ends and confer end protection. Here we show that HAATI arises when telomere loss triggers a newly recognized illegitimate translocation pathway that requires RNAi factors. While RNAi is necessary for the translocation events that mobilize ribosomal DNA (rDNA) tracts to all chromosome ends (forming "HAATIrDNA" chromosomes), it is dispensable for HAATIrDNA maintenance. Surprisingly, Dicer (Dcr1) plays a separate, RNAi-independent role in preventing formation of the rare HAATI subtype in which a different repetitive element (the subtelomeric element) replaces telomeres. Using genetics and fusions between shelterin components and rDNA-binding proteins, we mapped the mechanism by which rDNA loci engage crucial end protection factors-despite the absence of telomere repeats-and secure end protection. Sequence analysis of HAATIrDNA genomes allowed us to propose RNA and DNA polymerase template-switching models for the mechanism of RNAi-triggered rDNA translocations. Collectively, our results reveal unforeseen roles for noncoding RNAs (ncRNAs) in assembling a telomere-free chromosome end protection device.


Subject(s)
DNA, Ribosomal , Heterochromatin , RNA Interference , Translocation, Genetic , DNA Repair , DNA-Binding Proteins/physiology , Rad51 Recombinase/physiology , Ribonuclease III/metabolism , Ribonuclease III/physiology , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/physiology , Shelterin Complex , Telomere , Telomere-Binding Proteins/metabolism , Terminal Repeat Sequences
15.
Sci Signal ; 11(513)2018 01 16.
Article in English | MEDLINE | ID: mdl-29339534

ABSTRACT

The transforming growth factor-ß (TGF-ß) and bone morphogenetic protein (BMP) family of cytokines critically regulates vascular morphogenesis and homeostasis. Impairment of TGF-ß or BMP signaling leads to heritable vascular disorders, including hereditary hemorrhagic telangiectasia (HHT). Drosha, a key enzyme for microRNA (miRNA) biogenesis, also regulates the TGF-ß and BMP pathway through interaction with Smads and their joint control of gene expression through miRNAs. We report that mice lacking Drosha in the vascular endothelium developed a vascular phenotype resembling HHT that included dilated and disorganized vasculature, arteriovenous fistulae, and hemorrhages. Exome sequencing of HHT patients who lacked known pathogenic mutations revealed an overrepresentation of rare nonsynonymous variants of DROSHA Two of these DROSHA variants (P100L and R279L) did not interact with Smads and were partially catalytically active. In zebrafish, expression of these mutants or morpholino-directed knockdown of Drosha resulted in angiogenesis defects and abnormal vascular permeability. Together, our studies point to an essential role of Drosha in vascular development and the maintenance of vascular integrity, and reveal a previously unappreciated link between Drosha dysfunction and HHT.


Subject(s)
Gene Expression Regulation , Mutation , Neovascularization, Pathologic , Ribonuclease III/genetics , Ribonuclease III/physiology , Telangiectasia, Hereditary Hemorrhagic/genetics , Animals , Case-Control Studies , Cells, Cultured , Child , Cohort Studies , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Male , Mice , Mice, Knockout , Morphogenesis , Pedigree , Phenotype , Ribonuclease III/metabolism , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic/metabolism , Telangiectasia, Hereditary Hemorrhagic/pathology , Zebrafish/embryology , Zebrafish/physiology
16.
Hepatology ; 67(6): 2414-2429, 2018 06.
Article in English | MEDLINE | ID: mdl-29091291

ABSTRACT

Fibrosis and cancer represent two major complications of chronic liver disease. MicroRNAs have been implicated in the development of fibrosis and cancer, thus constituting potential therapeutic targets. Here, we investigated the role of microRNA-21 (miR-21), a microRNA that has been implicated in the development of fibrosis in multiple organs and has also been suggested to act as an "oncomir." Accordingly, miR-21 was the microRNA that showed the strongest up-regulation in activated hepatic stellate cells (HSCs) in multiple models of fibrogenesis, with an 8-fold to 24-fold induction compared to quiescent HSCs. However, miR-21 antisense inhibition did not suppress the activation of murine or human HSCs in culture or in liver slices. Moreover, genetic deletion of miR-21 in two independently generated knockout mice or miR-21 antisense inhibition did not alter HSC activation or liver fibrosis in models of toxic and biliary liver injury. Despite a strong up-regulation of miR-21 in injury-associated hepatocellular carcinoma and in cholangiocarcinoma, miR-21 deletion or antisense inhibition did not reduce the development of liver tumors. As inhibition of the most up-regulated microRNA did not affect HSC activation, liver fibrosis, or fibrosis-associated liver cancer, we additionally tested the role of microRNAs in HSCs by HSC-specific Dicer deletion. Although Dicer deletion decreased microRNA expression in HSCs and altered the expression of select genes, it only exerted negligible effects on HSC activation and liver fibrosis. CONCLUSION: Genetic and pharmacologic manipulation of miR-21 does not inhibit the development of liver fibrosis and liver cancer. Moreover, suppression of microRNA synthesis does not significantly affect HSC phenotype and activation. (Hepatology 2018;67:2414-2429).


Subject(s)
DEAD-box RNA Helicases/physiology , Hepatic Stellate Cells/physiology , Liver Cirrhosis/etiology , MicroRNAs/physiology , Ribonuclease III/physiology , Animals , Female , Humans , Male , Mice, Knockout
17.
RNA ; 24(1): 18-29, 2018 01.
Article in English | MEDLINE | ID: mdl-29079634

ABSTRACT

Dicer-dependent small noncoding RNAs play important roles in gene regulation in a wide variety of organisms. Endogenous small interfering RNAs (siRNAs) are part of an ancient pathway of transposon control in plants and animals. The ciliate, Oxytricha trifallax, has approximately 16,000 gene-sized chromosomes in its somatic nucleus. Long noncoding RNAs establish high ploidy levels at the onset of sexual development, but the factors that regulate chromosome copy numbers during cell division and growth have been a mystery. We report a novel function of a class of Dicer (Dcl-1)- and RNA-dependent RNA polymerase (RdRP)-dependent endogenous small RNAs in regulating chromosome copy number and gene dosage in O. trifallax Asexually growing populations express an abundant class of 21-nt sRNAs that map to both coding and noncoding regions of most chromosomes. These sRNAs are bound to chromatin and their levels surprisingly do not correlate with mRNA levels. Instead, the levels of these small RNAs correlate with genomic DNA copy number. Reduced sRNA levels in dcl-1 or rdrp mutants lead to concomitant reduction in chromosome copy number. Furthermore, these cells show no signs of transposon activation, but instead display irregular nuclear architecture and signs of replication stress. In conclusion, Oxytricha Dcl-1 and RdRP-dependent small RNAs that derive from the somatic nucleus contribute to the maintenance of gene dosage, possibly via a role in DNA replication, offering a novel role for these small RNAs in eukaryotes.


Subject(s)
DNA, Protozoan/genetics , Oxytricha/genetics , RNA, Protozoan/physiology , RNA, Small Untranslated/physiology , Chromosomes/genetics , DNA Copy Number Variations , DNA Replication , Epigenesis, Genetic , Protozoan Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease III/physiology
18.
Biochim Biophys Acta Gene Regul Mech ; 1860(12): 1179-1188, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29109067

ABSTRACT

microRNAs (miRNAs) are first transcribed as long, primary transcripts, which are then processed by multiple enzymes and proteins to generate the single-stranded, approximately 22-nucleotide (nt)-long mature miRNAs. A critical step in animal miRNA biogenesis is the cleavage of primary miRNA transcripts (pri-miRNAs) to produce precursor miRNAs (pre-miRNAs) by the enzyme Drosha. How Drosha recognizes its substrates remains incompletely understood. In this study we constructed a series of human Drosha mutants and examined their enzymatic activities and interaction with RNAs. We found that the N-terminal region is required for the nuclear localization and cellular function of Drosha. And in contrast to previous reports, we showed that the double-stranded RNA binding domain (RBD) of Drosha exhibited a weak but noticeable affinity for RNA. Compared to the RBDs of other RNA-binding proteins, the RBD of Drosha has a short insert, whose mutations reduced RNA binding and pri-miRNA cleavage. Overexpression of Drosha RBD mutants in a reporter assay corroborated their deficiencies in Drosha activity in cell cultures. In addition, we found that point mutations in the RNaseIIIb domain of Drosha implicated in Wilms tumors differentially affected cleavage of the 5' and 3' strands of pri-miRNAs in vitro. In conclusion, our results provided important insights into the mechanism of pri-miRNA processing by human Drosha.


Subject(s)
Double-Stranded RNA Binding Motif/physiology , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Ribonuclease III/physiology , Cells, Cultured , Humans , Mutation , RNA/metabolism , Ribonuclease III/chemistry
19.
Nucleic Acids Res ; 45(18): 10369-10379, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-28977573

ABSTRACT

MicroRNAs (miRNAs) play a pivotal role in the regulation of cellular gene expression via the conserved RNA interference (RNAi) mechanism. Biogenesis of the unusual miR-451 does not require Dicer. This molecule is instead processed by the Argonaute 2 (Ago2) enzyme. Similarly, unconventional short hairpin RNA (shRNA) molecules have been designed as miR-451 mimics that rely exclusively on Ago2 for maturation. We will review recent progress made in the understanding of this alternative processing route. Next, we describe different Dicer-independent shRNA designs that have been developed and discuss their therapeutic advantages and disadvantages. As an example, we will present the route towards development of a durable gene therapy against HIV-1.


Subject(s)
DEAD-box RNA Helicases/physiology , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , RNA, Double-Stranded/metabolism , Ribonuclease III/physiology , Animals , Humans , MicroRNAs/chemistry , Nucleic Acid Conformation , RNA Interference/physiology , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism
20.
Nucleic Acids Res ; 45(16): 9398-9412, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28934503

ABSTRACT

In mammals, faithful inheritance of genomic methylation patterns ensures proper gene regulation and cell behaviour, impacting normal development and fertility. Following establishment, genomic methylation patterns are transmitted through S-phase by the maintenance methyltransferase Dnmt1. Using a protein interaction screen, we identify Microprocessor component DROSHA as a novel DNMT1-interactor. Drosha-deficient embryonic stem (ES) cells display genomic hypomethylation that is not accounted for by changes in the levels of DNMT proteins. DNMT1-mediated methyltransferase activity is also reduced in these cells. We identify two transcripts that are specifically upregulated in Drosha- but not Dicer-deficient ES cells. Regions within these transcripts predicted to form stem-loop structures are processed by Microprocessor and can inhibit DNMT1-mediated methylation in vitro. Our results highlight DROSHA as a novel regulator of mammalian DNA methylation and we propose that DROSHA-mediated processing of RNA is necessary to ensure full DNMT1 activity. This adds to the DROSHA repertoire of non-miRNA dependent functions as well as implicating RNA in regulating DNMT1 activity and correct levels of genomic methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Ribonuclease III/physiology , Animals , CRISPR-Cas Systems , Cells, Cultured , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , Embryonic Stem Cells/enzymology , HEK293 Cells , Humans , Mice , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...