Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-38014251

ABSTRACT

Hypertrophic cardiomyopathy (HCM) results from pathogenic variants in sarcomeric protein genes, that increase myocyte energy demand and lead to cardiac hypertrophy. But it is unknown whether a common metabolic trait underlies the cardiac phenotype at early disease stage. This study characterized two HCM mouse models (R92W-TnT, R403Q-MyHC) that demonstrate differences in mitochondrial function at early disease stage. Using a combination of cardiac phenotyping, transcriptomics, mass spectrometry-based metabolomics and computational modeling, we discovered allele-specific differences in cardiac structure/function and metabolic changes. TnT-mutant hearts had impaired energy substrate metabolism and increased phospholipid remodeling compared to MyHC-mutants. TnT-mutants showed increased incorporation of saturated fatty acid residues into ceramides, cardiolipin, and increased lipid peroxidation, that could underlie allele-specific differences in mitochondrial function and cardiomyopathy.

2.
Sci Rep ; 11(1): 13163, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162896

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is characterized by phenotypic heterogeneity. We investigated the molecular basis of the cardiac phenotype in two mouse models at established disease stage (mouse-HCM), and human myectomy tissue (human-HCM). We analyzed the transcriptome in 2 mouse models with non-obstructive HCM (R403Q-MyHC, R92W-TnT)/littermate-control hearts at 24 weeks of age, and in myectomy tissue of patients with obstructive HCM/control hearts (GSE36961, GSE36946). Additionally, we examined myocyte redox, cardiac mitochondrial DNA copy number (mtDNA-CN), mt-respiration, mt-ROS generation/scavenging and mt-Ca2+ handling in mice. We identified distinct allele-specific gene expression in mouse-HCM, and marked differences between mouse-HCM and human-HCM. Only two genes (CASQ1, GPT1) were similarly dysregulated in both mutant mice and human-HCM. No signaling pathway or transcription factor was predicted to be similarly dysregulated (by Ingenuity Pathway Analysis) in both mutant mice and human-HCM. Losartan was a predicted therapy only in TnT-mutant mice. KEGG pathway analysis revealed enrichment for several metabolic pathways, but only pyruvate metabolism was enriched in both mutant mice and human-HCM. Both mutant mouse myocytes demonstrated evidence of an oxidized redox environment. Mitochondrial complex I RCR was lower in both mutant mice compared to controls. MyHC-mutant mice had similar mtDNA-CN and mt-Ca2+ handling, but TnT-mutant mice exhibited lower mtDNA-CN and impaired mt-Ca2+ handling, compared to littermate-controls. Molecular profiling reveals differences in gene expression, transcriptional regulation, intracellular signaling and mt-number/function in 2 mouse models at established disease stage. Further studies are needed to confirm differences in gene expression between mouse and human-HCM, and to examine whether cardiac phenotype, genotype and/or species differences underlie the divergence in molecular profiles.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Transcriptome , Animals , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/diagnostic imaging , Cardiomyopathy, Hypertrophic/metabolism , Carrier Proteins/genetics , Disease Models, Animal , Echocardiography , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Mice, Transgenic , Mitochondria, Heart/metabolism , Mutation, Missense , Myocardium/metabolism , Myosin Heavy Chains/genetics , Phenotype , Point Mutation , RNA, Messenger/genetics , Species Specificity , Troponin T/genetics , Ventricular Outflow Obstruction/etiology , Ventricular Outflow Obstruction/genetics
3.
PLoS Genet ; 17(6): e1009655, 2021 06.
Article in English | MEDLINE | ID: mdl-34181646

ABSTRACT

During spermatogenesis, the process in which sperm for fertilization are produced from germline cells, gene expression is spatiotemporally highly regulated. In Drosophila, successful expression of extremely large male fertility factor genes on Y-chromosome spanning some megabases due to their gigantic intron sizes is crucial for spermatogenesis. Expression of such extremely large genes must be challenging, but the molecular mechanism that allows it remains unknown. Here we report that a novel RNA-binding protein Maca, which contains two RNA-recognition motifs, is crucial for this process. maca null mutant male flies exhibited a failure in the spermatid individualization process during spermatogenesis, lacked mature sperm, and were completely sterile, while maca mutant female flies were fully fertile. Proteomics and transcriptome analyses revealed that both protein and mRNA abundance of the gigantic male fertility factor genes kl-2, kl-3, and kl-5 (kl genes) are significantly decreased, where the decreases of kl-2 are particularly dramatic, in maca mutant testes. Splicing of the kl-3 transcripts was also dysregulated in maca mutant testes. All these physiological and molecular phenotypes were rescued by a maca transgene in the maca mutant background. Furthermore, we found that in the control genetic background, Maca is exclusively expressed in spermatocytes in testes and enriched at Y-loop A/C in the nucleus, where the kl-5 primary transcripts are localized. Our data suggest that Maca increases transcription processivity, promotes successful splicing of gigantic introns, and/or protects transcripts from premature degradation, of the kl genes. Our study identified a novel RNA-binding protein Maca that is crucial for successful expression of the gigantic male fertility factor genes, spermatogenesis, and male fertility.


Subject(s)
Drosophila melanogaster/genetics , RNA-Binding Proteins/genetics , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Transcriptome , Animals , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Female , Fertility/genetics , Gene Expression Regulation , Gene Ontology , Genes, Reporter , Genetic Complementation Test , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Molecular Sequence Annotation , Mutation , RNA-Binding Proteins/metabolism , Spermatids/cytology , Spermatids/growth & development , Spermatocytes/cytology , Spermatocytes/growth & development , Testis/cytology , Testis/metabolism , Y Chromosome/chemistry
4.
RNA Biol ; 16(10): 1386-1400, 2019 10.
Article in English | MEDLINE | ID: mdl-31195914

ABSTRACT

Metamorphosis is an intricate developmental process in which large-scale remodelling of mRNA and microRNA (miRNA) profiles leads to orchestrated tissue remodelling and organogenesis. Whether, which, and how, ribonucleases (RNases) are involved in the RNA profile remodelling during metamorphosis remain unknown. Human Regnase-1 (also known as MCPIP1 and Zc3h12a) RNase remodels RNA profile by cleaving specific RNAs and is a crucial modulator of immune-inflammatory and cellular defence. Here, we studied Drosophila CG10889, which we named Drosophila Regnase-1, an ortholog of human Regnase-1. The larva-to-adult metamorphosis in Drosophila includes two major transitions, larva-to-pupa and pupa-to-adult. regnase-1 knockout flies developed until the pupa stage but could not complete pupa-to-adult transition, dying in puparium case. Regnase-1 RNase activity is required for completion of pupa-to-adult transition as transgenic expression of wild-type Drosophila Regnase-1, but not the RNase catalytic-dead mutants, rescued the pupa-to-adult transition in regnase-1 knockout. High-throughput RNA sequencing revealed that regnase-1 knockout flies fail to remodel mRNA and miRNA profiles during the larva-to-pupa transition. Thus, we uncovered the roles of Drosophila Regnase-1 in the larva-to-adult metamorphosis and large-scale remodelling of mRNA and miRNA profiles during this metamorphosis process.


Subject(s)
Drosophila/physiology , Metamorphosis, Biological/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Ribonucleases/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Catalysis , Gene Expression , Hydrolysis , Larva , Mutation , Ribonucleases/genetics
5.
PLoS One ; 14(5): e0217603, 2019.
Article in English | MEDLINE | ID: mdl-31145769

ABSTRACT

RNA methyltransferases post-transcriptionally add methyl groups to RNAs, which can regulate their fates and functions. Human BCDIN3D (Bicoid interacting 3 domain containing RNA methyltransferase) has been reported to specifically methylate the 5'-monophosphates of pre-miR-145 and cytoplasmic tRNAHis. Methylation of the 5'-monophosphate of pre-miR-145 blocks its cleavage by the miRNA generating enzyme Dicer, preventing generation of miR-145. Elevated expression of BCDIN3D has been associated with poor prognosis in breast cancer. However, the biological functions of BCDIN3D and its orthologs remain unknown. Here we studied the biological and molecular functions of CG1239, a Drosophila ortholog of BCDIN3D. We found that ovary-specific knockdown of Drosophila BCDIN3D causes female sterility. High-throughput sequencing revealed that miRNA and mRNA profiles are dysregulated in BCDIN3D knockdown ovaries. Pathway analysis showed that many of the dysregulated genes are involved in metabolic processes, ribonucleoprotein complex regulation, and translational control. Our results reveal BCDIN3D's biological role in female fertility and its molecular role in defining miRNA and mRNA profiles in ovaries.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Fertility/genetics , Methyltransferases/genetics , Ovary/growth & development , Protein O-Methyltransferase/genetics , Animals , DNA-Binding Proteins/genetics , Drosophila melanogaster/growth & development , Female , Gene Expression Regulation, Developmental/genetics , Metabolism/genetics , Methylation , MicroRNAs/genetics , Ovary/metabolism , RNA, Messenger/genetics , Ribonuclease III/genetics
6.
RNA ; 25(7): 825-839, 2019 07.
Article in English | MEDLINE | ID: mdl-30979781

ABSTRACT

Drosophila Belle (human ortholog DDX3) is a conserved DEAD-box RNA helicase implicated in regulating gene expression. However, the molecular mechanisms by which Belle/DDX3 regulates gene expression are poorly understood. Here we performed systematic mutational analysis to determine the contributions of conserved motifs within Belle to its in vivo function. We found that Belle RNA-binding and RNA-unwinding activities and intrinsically disordered regions (IDRs) are required for Belle in vivo function. Expression of Belle ATPase mutants that cannot bind, hydrolyze, or release ATP resulted in dominant toxic phenotypes. Mechanistically, we discovered that Belle up-regulates reporter protein level when tethered to reporter mRNA, without corresponding changes at the mRNA level, indicating that Belle promotes translation of mRNA that it binds. Belle ATPase activity and amino-terminal IDR were required for this translational promotion activity. We also found that ectopic ovary expression of dominant Belle ATPase mutants decreases levels of cyclin proteins, including Cyclin B, without corresponding changes in their mRNA levels. Finally, we found that Belle binds endogenous cyclin B mRNA. We propose that Belle promotes translation of specific target mRNAs, including cyclin B mRNA, in an ATPase activity-dependent manner.


Subject(s)
Adenosine Triphosphatases/metabolism , DEAD-box RNA Helicases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intrinsically Disordered Proteins/metabolism , RNA Helicases/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Adenosine Triphosphatases/genetics , Animals , Cyclin B/genetics , Cyclin B/metabolism , DEAD-box RNA Helicases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Eye/growth & development , Eye/metabolism , Female , Infertility, Female , Infertility, Male , Intrinsically Disordered Proteins/genetics , Male , Phenotype , RNA Helicases/genetics , RNA, Messenger/genetics
7.
Nat Commun ; 10(1): 1074, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824694

ABSTRACT

The originally published version of this Article contained an error in Figure 1a, in which the length of the protein fragment produced by the MARF1 null allele was incorrectly labelled as '34aa' rather than the corrected '103aa'.Also, the second sentence of the third paragraph of the Results originally read 'The MARF1null allele has a 241-nt-long deletion introduced at proximal to the N-terminal end of the protein, which produced a premature stop codon, resulting in production of the N-terminal 34 aa fragment of MARF1 (Fig. 1a).' In the corrected version, '34aa' is replaced by '103aa'.These errors have now been corrected in both the PDF and the HTML versions of the Article.

8.
Front Cardiovasc Med ; 6: 170, 2019.
Article in English | MEDLINE | ID: mdl-31921893

ABSTRACT

Background: Hypertrophic cardiomyopathy (HCM) is characterized by myocyte hypertrophy and fibrosis. Studies in two mouse models (R92W-TnT/R403Q-MyHC) at early HCM stage revealed upregulation of endothelin (ET1) signaling in both mutants, but TGFß signaling only in TnT mutants. Dysregulation of miR-29 expression has been implicated in cardiac fibrosis. But it is unknown whether expression of miR-29a/b/c and profibrotic genes is commonly regulated in mouse and human HCM. Methods: In order to understand mechanisms underlying fibrosis in HCM, and examine similarities/differences in expression of miR-29a/b/c and several profibrotic genes in mouse and human HCM, we performed parallel studies in rat cardiac myocyte/fibroblast cultures, examined gene expression in two mouse models of (non-obstructive) HCM (R92W-TnT, R403Q-MyHC)/controls at early (5 weeks) and established (24 weeks) disease stage, and analyzed publicly available mRNA/miRNA expression data from obstructive-HCM patients undergoing septal myectomy/controls (unused donor hearts). Results: Myocyte cultures: ET1 increased superoxide/H2O2, stimulated TGFß expression/secretion, and suppressed miR-29a expression in myocytes. The effect of ET1 on miR-29 and TGFß expression/secretion was antagonized by N-acetyl-cysteine, a reactive oxygen species scavenger. Fibroblast cultures: ET1 had no effect on pro-fibrotic gene expression in fibroblasts. TGFß1/TGFß2 suppressed miR-29a and increased collagen expression, which was abolished by miR-29a overexpression. Mouse and human HCM: Expression of miR-29a/b/c was lower, and TGFB1/collagen gene expression was higher in TnT mutant-LV at 5 and 24 weeks; no difference was observed in expression of these genes in MyHC mutant-LV and in human myectomy tissue. TGFB2 expression was higher in LV of both mutant mice and human myectomy tissue. ACE2, a negative regulator of the renin-angiotensin-aldosterone system, was the most upregulated transcript in human myectomy tissue. Pathway analysis predicted upregulation of the anti-hypertrophic/anti-fibrotic liver X receptor/retinoid X receptor (LXR/RXR) pathway only in human myectomy tissue. Conclusions: Our in vitro studies suggest that activation of ET1 signaling in cardiac myocytes increases reactive oxygen species and stimulates TGFß secretion, which downregulates miR-29a and increases collagen in fibroblasts, thus contributing to fibrosis. Our gene expression studies in mouse and human HCM reveal allele-specific differences in miR-29 family/profibrotic gene expression in mouse HCM, and activation of anti-hypertrophic/anti-fibrotic genes and pathways in human HCM.

9.
Nat Commun ; 9(1): 4031, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279526

ABSTRACT

Post-transcriptional regulation of gene expression plays an essential role during oocyte maturation. Here we report that Drosophila MARF1 (Meiosis Regulator And mRNA Stability Factor 1), which consists of one RNA-recognition motif and six tandem LOTUS domains with unknown molecular function, is essential for oocyte maturation. When tethered to a reporter mRNA, MARF1 post-transcriptionally silences reporter expression by shortening reporter mRNA poly-A tail length and thereby reducing reporter protein level. This activity is mediated by the MARF1 LOTUS domain, which binds the CCR4-NOT deadenylase complex. MARF1 binds cyclin A mRNA and shortens its poly-A tail to reduce Cyclin A protein level during oocyte maturation. This study identifies MARF1 as a regulator in oocyte maturation and defines the conserved LOTUS domain as a post-transcriptional effector domain that recruits CCR4-NOT deadenylase complex to shorten target mRNA poly-A tails and suppress their translation.


Subject(s)
Cyclin A/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Oocytes/metabolism , RNA 3' End Processing , Ribonucleases/metabolism , Animals , Animals, Genetically Modified , Drosophila , Drosophila Proteins/genetics , Female , Fertility , Male , Meiosis , Membrane Proteins/genetics , Protein Transport
10.
Heliyon ; 4(7): e00706, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30094376

ABSTRACT

Drosophila Blanks is a testes-specific RNA-binding protein required for post-meiotic spermiogenesis. However, Blanks's role in regulating RNA populations in the testes remains unknown. We performed small RNA and mRNA high-throughput sequencing in blanks mutant testes and controls. We identified two miRNAs, one siRNA, and hundreds of mRNAs that are significantly upregulated or downregulated in blanks mutant testes. Pathway analysis revealed that differentially expressed mRNAs are involved in catabolic and metabolic processes, anion and cation transport, mating, and reproductive behavior. Our results reveal that Blanks plays important roles in defining testicular small RNA and mRNA profiles.

11.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563334

ABSTRACT

Hypertrophic cardiomyopathy (HCM) stems from mutations in sarcomeric proteins that elicit distinct biophysical sequelae, which in turn may yield radically different intracellular signaling and molecular pathologic profiles. These signaling events remain largely unaddressed by clinical trials that have selected patients based on clinical HCM diagnosis, irrespective of genotype. In this study, we determined how two mouse models of HCM differ, with respect to cellular/mitochondrial function and molecular biosignatures, at an early stage of disease. We show that hearts from young R92W-TnT and R403Q-αMyHC mutation-bearing mice differ in their transcriptome, miRNome, intracellular redox environment, mitochondrial antioxidant defense mechanisms, and susceptibility to mitochondrial permeability transition pore opening. Pathway analysis of mRNA-sequencing data and microRNA profiles indicate that R92W-TnT mutants exhibit a biosignature consistent with activation of profibrotic TGF-ß signaling. Our results suggest that the oxidative environment and mitochondrial impairment in young R92W-TnT mice promote activation of TGF-ß signaling that foreshadows a pernicious phenotype in young individuals. Of the two mutations, R92W-TnT is more likely to benefit from anti-TGF-ß signaling effects conferred by angiotensin receptor blockers and may be responsive to mitochondrial antioxidant strategies in the early stage of disease. Molecular and functional profiling may therefore serve as aids to guide precision therapy for HCM.


Subject(s)
Alleles , Cardiomyopathy, Hypertrophic/metabolism , MicroRNAs/metabolism , Mitochondria/metabolism , Transcriptome , Animals , Antioxidants , Calcium/metabolism , Cardiomyopathy, Hypertrophic/genetics , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Mitochondria/genetics , Muscle Cells/metabolism , Mutation , Permeability , Phenotype , RNA, Messenger/metabolism , Sequence Analysis, RNA , Signal Transduction
12.
Biochem Biophys Res Commun ; 498(4): 1022-1027, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29550490

ABSTRACT

Drosophila Dicer-2 processes RNA substrates into short interfering RNAs (siRNAs). Loquacious-PD (Loqs-PD), a dsRNA-binding protein that associates with Dicer-2, is required for processing of a subset of RNA substrates including hairpin RNAs into siRNAs. Inorganic phosphate-a small molecule present in all cell types-inhibits Dicer-2 from processing precursor of microRNAs (pre-miRNAs), which are processed by Dicer-1. Whether or how Loqs-PD modulates the inhibitory effect of inorganic phosphate on Dicer-2 processing of RNA substrates is unknown. To address this question, I performed in vitro hairpin RNA processing assay with Dicer-2 in the presence or absence of Loqs-PD and/or inorganic phosphate. I found that inorganic phosphate inhibits Dicer-2 alone, but not Dicer-2 + Loqs-PD, from processing blunt-end hairpin RNAs into siRNAs. Thus, Loqs-PD removes the inhibitory effect of inorganic phosphate on Dicer-2 processing of blunt-end hairpin RNAs, allowing siRNA production in the presence of inorganic phosphate.


Subject(s)
Drosophila Proteins/metabolism , Phosphates/pharmacology , RNA Helicases/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins/physiology , Ribonuclease III/metabolism , Animals , Drosophila , Drosophila Proteins/antagonists & inhibitors , MicroRNAs/metabolism , RNA Helicases/antagonists & inhibitors , Ribonuclease III/antagonists & inhibitors
13.
Nucleic Acids Res ; 46(7): 3726-3741, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29373753

ABSTRACT

Dicer partner proteins Drosophila Loquacious-PB (Loqs-PB) and human TRBP tune the length of miRNAs produced by Dicer from a subset of pre-miRNAs and thereby alter their target repertoire, by an unknown mechanism. Here, we developed a novel high-throughput method that we named Dram-seq (Dice randomized pre-miRNA pool and seq) to study length distributions of miRNAs produced from thousands of different pre-miRNA variants. Using Dram-seq, we found that a base-mismatch in the pre-miRNA stem can alter the length of miRNAs compared with a base-pair at the same position in both Drosophila and human, and is important for the miRNA length tuning by Loqs-PB. Loqs-PB directly bound base-mismatched nucleotides in the pre-miRNA stem. We speculate that Loqs-PB tunes miRNA length by changing the conformation of base-mismatched nucleotides in the pre-miRNA stem to that of base-paired ones and thereby altering the distance of the pre-miRNA stem.


Subject(s)
Drosophila Proteins/genetics , Homeodomain Proteins/genetics , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Animals , Base Pairing/genetics , DEAD-box RNA Helicases/genetics , Drosophila melanogaster/genetics , Humans , Nucleic Acid Conformation , Ribonuclease III/genetics
14.
Methods Mol Biol ; 1680: 101-121, 2018.
Article in English | MEDLINE | ID: mdl-29030844

ABSTRACT

Dicer enzymes produce small silencing RNAs such as microRNAs (miRNAs) and small interfering RNAs (siRNAs), which then are loaded into Argonaute proteins and act as sequence-specific guides. A powerful tool to understand the molecular mechanism of small silencing RNA production by Dicers is an in vitro RNA processing assay using recombinant Dicer proteins. Such biochemical analyses have elucidated the substrate specificities and kinetics of Dicers, the mechanism by which the length of small RNAs produced by Dicers is determined, and the effects of Dicer-partner proteins and endogenous small molecules such as ATP and inorganic phosphate on small RNA production by Dicers, among others. Here, we describe methods for in vitro small RNA production assay using recombinant human and Drosophila Dicer proteins.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , RNA Interference , RNA, Small Interfering/genetics , Ribonuclease III/metabolism , Animals , Humans , In Vitro Techniques , Isotope Labeling , Kinetics , RNA, Small Interfering/metabolism , Recombinant Proteins
15.
RNA ; 23(7): 1139-1153, 2017 07.
Article in English | MEDLINE | ID: mdl-28416567

ABSTRACT

Drosophila Dicer-2 efficiently and precisely produces 21-nucleotide (nt) siRNAs from long double-stranded RNA (dsRNA) substrates and loads these siRNAs onto the effector protein Argonaute2 for RNA silencing. The functional roles of each domain of the multidomain Dicer-2 enzyme in the production and loading of siRNAs are not fully understood. Here we characterized Dicer-2 mutants lacking either the N-terminal helicase domain or the C-terminal dsRNA-binding domain (CdsRBD) (ΔHelicase and ΔCdsRBD, respectively) in vivo and in vitro. We found that ΔCdsRBD Dicer-2 produces siRNAs with lowered efficiency and length fidelity, producing a smaller ratio of 21-nt siRNAs and higher ratios of 20- and 22-nt siRNAs in vivo and in vitro. We also found that ΔCdsRBD Dicer-2 cannot load siRNA duplexes to Argonaute2 in vitro. Consistent with these findings, we found that ΔCdsRBD Dicer-2 causes partial loss of RNA silencing activity in vivo. Thus, Dicer-2 CdsRBD is crucial for the efficiency and length fidelity in siRNA production and for siRNA loading. Together with our previously published findings, we propose that CdsRBD binds the proximal body region of a long dsRNA substrate whose 5'-monophosphate end is anchored by the phosphate-binding pocket in the PAZ domain. CdsRBD aligns the RNA to the RNA cleavage active site in the RNase III domain for efficient and high-fidelity siRNA production. This study reveals multifunctions of Dicer-2 CdsRBD and sheds light on the molecular mechanism by which Dicer-2 produces 21-nt siRNAs with a high efficiency and fidelity for efficient RNA silencing.


Subject(s)
Argonaute Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , RNA Helicases/chemistry , RNA Helicases/metabolism , RNA, Small Interfering/metabolism , Ribonuclease III/chemistry , Ribonuclease III/metabolism , Animals , Animals, Genetically Modified , Binding Sites , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , In Vitro Techniques , Mutation , Protein Binding , Protein Domains , RNA Helicases/genetics , Ribonuclease III/genetics
16.
Proc Natl Acad Sci U S A ; 113(49): 14031-14036, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27872309

ABSTRACT

The enzyme Dicer produces small silencing RNAs such as micro-RNAs (miRNAs) and small interfering RNAs (siRNAs). In Drosophila, Dicer-1 produces ∼22-24-nt miRNAs from pre-miRNAs, whereas Dicer-2 makes 21-nt siRNAs from long double-stranded RNAs (dsRNAs). How Dicer-2 precisely makes 21-nt siRNAs with a remarkably high fidelity is unknown. Here we report that recognition of the 5'-monophosphate of a long dsRNA substrate by a phosphate-binding pocket in the Dicer-2 PAZ (Piwi, Argonaute, and Zwille/Pinhead) domain is crucial for the length fidelity, but not the efficiency, in 21-nt siRNA production. Loss of the length fidelity, meaning increased length heterogeneity of siRNAs, caused by point mutations in the phosphate-binding pocket of the Dicer-2 PAZ domain decreased RNA silencing activity in vivo, showing the importance of the high fidelity to make 21-nt siRNAs. We propose that the 5'-monophosphate of a long dsRNA substrate is anchored by the phosphate-binding pocket in the Dicer-2 PAZ domain and the distance between the pocket and the RNA cleavage active site in the RNaseIII domain corresponds to the 21-nt pitch in the A-form duplex of a long dsRNA substrate, resulting in high-fidelity 21-nt siRNA production. This study sheds light on the molecular mechanism by which Dicer-2 produces 21-nt siRNAs with a remarkably high fidelity for efficient RNA silencing.


Subject(s)
Drosophila Proteins/physiology , RNA Helicases/physiology , RNA Interference/physiology , RNA, Small Interfering/biosynthesis , Ribonuclease III/physiology , Amino Acid Sequence , Animals , Binding Sites , Drosophila Proteins/metabolism , Drosophila melanogaster , MicroRNAs , Phosphate-Binding Proteins/metabolism , Protein Binding , Protein Domains , RNA Helicases/metabolism , RNA, Double-Stranded/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonuclease III/metabolism , Substrate Specificity
17.
PLoS One ; 11(5): e0156065, 2016.
Article in English | MEDLINE | ID: mdl-27196440

ABSTRACT

MicroRNAs (miRNAs) are recognized as important regulators of cardiac development, hypertrophy and fibrosis. Recent studies have demonstrated that genetic variations which cause alterations in miRNA:target interactions can lead to disease. We hypothesized that genetic variations in miRNAs that regulate cardiac hypertrophy/fibrosis might be involved in generation of the cardiac phenotype in patients diagnosed with hypertrophic cardiomyopathy (HCM). To investigate this question, we Sanger sequenced 18 miRNA genes previously implicated in myocyte hypertrophy/fibrosis and apoptosis, using genomic DNA isolated from the leukocytes of 199 HCM patients. We identified a single nucleotide polymorphism (rs6971711, C57T SNP) at the 17th position of mature miR-590-3p (= 57th position of pre-miR-590) that is common in individuals of African ancestry. SNP frequency was higher in African American HCM patients (n = 55) than ethnically-matched controls (n = 100), but the difference was not statistically significant (8.2% vs. 6.5%; p = 0.5). Using a cell culture system, we discovered that presence of this SNP resulted in markedly lower levels of mature miR-590-5p (39 ± 16%, p<0.003) and miR-590-3p (20 ± 2%, p<0.003), when compared with wild-type (WT) miR-590, without affecting levels of pri-miR-590 and pre-miR-590. Consistent with this finding, the SNP resulted in reduced target suppression when compared to WT miR-590 (71% suppression by WT vs 60% suppression by SNP, p<0.03). Since miR-590 can regulate TGF-ß, Activin A and Akt signaling, SNP-induced reduction in miR-590 biogenesis could influence cardiac phenotype by de-repression of these signaling pathways. Since the SNP is only present in African Americans, population studies in this patient population would be valuable to investigate effects of this SNP on myocyte function and cardiac physiology.


Subject(s)
Black or African American/genetics , Cardiomyopathy, Hypertrophic/genetics , MicroRNAs/genetics , Polymorphism, Single Nucleotide , Cardiomyopathy, Hypertrophic/ethnology , Cardiomyopathy, Hypertrophic/pathology , Case-Control Studies , HEK293 Cells , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
18.
J Struct Funct Genomics ; 16(1): 25-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25618148

ABSTRACT

The putative translation elongation factor Mbar_A0971 from the methanogenic archaeon Methanosarcina barkeri was proposed to be the pyrrolysine-specific paralogue of EF-Tu ("EF-Pyl"). In the present study, the crystal structures of its homologue from Methanosarcina mazei (MM1309) were determined in the GMPPNP-bound, GDP-bound, and apo forms, by the single-wavelength anomalous dispersion phasing method. The three MM1309 structures are quite similar (r.m.s.d. < 0.1 Å). The three domains, corresponding to domains 1, 2, and 3 of EF-Tu/SelB/aIF2γ, are packed against one another to form a closed architecture. The MM1309 structures resemble those of bacterial/archaeal SelB, bacterial EF-Tu in the GTP-bound form, and archaeal initiation factor aIF2γ, in this order. The GMPPNP and GDP molecules are visible in their co-crystal structures. Isothermal titration calorimetry measurements of MM1309·GTP·Mg(2+), MM1309·GDP·Mg(2+), and MM1309·GMPPNP·Mg(2+) provided dissociation constants of 0.43, 26.2, and 222.2 µM, respectively. Therefore, the affinities of MM1309 for GTP and GDP are similar to those of SelB rather than those of EF-Tu. Furthermore, the switch I and II regions of MM1309 are involved in domain-domain interactions, rather than nucleotide binding. The putative binding pocket for the aminoacyl moiety on MM1309 is too small to accommodate the pyrrolysyl moiety, based on a comparison of the present MM1309 structures with that of the EF-Tu·GMPPNP·aminoacyl-tRNA ternary complex. A hydrolysis protection assay revealed that MM1309 binds cysteinyl (Cys)-tRNA(Cys) and protects the aminoacyl bond from non-enzymatic hydrolysis. Therefore, we propose that MM1309 functions as either a guardian protein that protects the Cys moiety from oxidation or an alternative translation factor for Cys-tRNA(Cys).


Subject(s)
Archaeal Proteins/chemistry , Guanosine Triphosphate/chemistry , Methanosarcina/chemistry , RNA, Transfer, Cys/chemistry , Amino Acid Sequence , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Calorimetry , Crystallography, X-Ray , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Guanylyl Imidodiphosphate/chemistry , Guanylyl Imidodiphosphate/metabolism , Kinetics , Methanosarcina/genetics , Methanosarcina/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Nucleic Acid Conformation , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Peptide Initiation Factors/chemistry , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Transfer, Cys/metabolism , Sequence Homology, Amino Acid
19.
Cell Cycle ; 13(11): 1671-6, 2014.
Article in English | MEDLINE | ID: mdl-24787225

ABSTRACT

The enzyme Dicer is central to the production of small silencing RNAs such as microRNAs (miRNAs) and small interfering RNAs (siRNAs). Like other insects, Drosophila melanogaster uses different Dicers to make siRNAs and miRNAs: Dicer-1 produces miRNAs from pre-miRNAs, whereas Dicer-2 generates siRNAs from long double-stranded RNA (dsRNA). How do the 2 Dicers achieve their substrate specificity? Here, we review recent findings that inorganic phosphate restricts the substrate specificity of Dicer-2 to long dsRNA. Inorganic phosphate inhibits Dicer-2 from binding and cleaving pre-miRNAs, without affecting the processing of long dsRNA. Crystal structures of a fragment of human Dicer in complex with an RNA duplex identify a phosphate-binding pocket that recognizes both the 5'-monophosphate of a substrate RNA and inorganic phosphate. We propose that inorganic phosphate occupies the phosphate-binding pocket in the fly Dicer-2, blocking binding of pre-miRNA and restricting pre-miRNA processing to Dicer-1. Thus, a small molecule can alter the substrate specificity of a nucleic acid-processing enzyme.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Models, Molecular , Organophosphates/pharmacology , RNA Helicases/metabolism , RNA, Small Untranslated/biosynthesis , Ribonuclease III/metabolism , Amino Acid Sequence , Animals , Base Pairing , DEAD-box RNA Helicases/chemistry , Drosophila Proteins/genetics , Gene Components/genetics , Humans , Molecular Sequence Data , RNA Helicases/genetics , RNA, Small Untranslated/chemistry , RNA, Small Untranslated/metabolism , Ribonuclease III/chemistry , Ribonuclease III/genetics , Substrate Specificity/drug effects , Substrate Specificity/genetics
20.
EMBO J ; 33(4): 371-84, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24488111

ABSTRACT

In Drosophila, Dicer-1 produces microRNAs (miRNAs) from pre-miRNAs, whereas Dicer-2 generates small interfering RNAs from long double-stranded RNA (dsRNA), a process that requires ATP hydrolysis. We previously showed that inorganic phosphate inhibits Dicer-2 cleavage of pre-miRNAs, but not long dsRNAs. Here, we report that phosphate-dependent substrate discrimination by Dicer-2 reflects dsRNA substrate length. Efficient processing by Dicer-2 of short dsRNA requires a 5' terminal phosphate and a two-nucleotide, 3' overhang, but does not require ATP. Phosphate inhibits cleavage of such short substrates. In contrast, cleavage of longer dsRNA requires ATP but no specific end structure: phosphate does not inhibit cleavage of these substrates. Mutation of a pair of conserved arginine residues in the Dicer-2 PAZ domain blocked cleavage of short, but not long, dsRNA. We propose that inorganic phosphate occupies a PAZ domain pocket required to bind the 5' terminal phosphate of short substrates, blocking their use and restricting pre-miRNA processing in flies to Dicer-1. Our study helps explain how a small molecule can alter the substrate specificity of a nucleic acid processing enzyme.


Subject(s)
Drosophila Proteins/drug effects , Drosophila melanogaster/metabolism , MicroRNAs/metabolism , Phosphates/pharmacology , RNA Helicases/drug effects , Ribonuclease III/drug effects , Amino Acid Substitution , Animals , Arginine , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Mutagenesis, Site-Directed , Protein Binding/drug effects , Protein Structure, Tertiary , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Double-Stranded/metabolism , Recombinant Fusion Proteins/metabolism , Ribonuclease III/chemistry , Ribonuclease III/genetics , Ribonuclease III/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...