Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters











Publication year range
1.
Viruses ; 13(6)2021 06 03.
Article in English | MEDLINE | ID: mdl-34205194

ABSTRACT

Mosquitoes are known as important vectors of many arthropod-borne (arbo)viruses causing disease in humans. These include dengue (DENV) and Zika (ZIKV) viruses. The exogenous small interfering (si)RNA (exo-siRNA) pathway is believed to be the main antiviral defense in arthropods, including mosquitoes. During infection, double-stranded RNAs that form during viral replication and infection are cleaved by the enzyme Dicer 2 (Dcr2) into virus-specific 21 nt vsiRNAs, which are subsequently loaded into Argonaute 2 (Ago2). Ago2 then targets and subsequently cleaves complementary RNA sequences, resulting in degradation of the target viral RNA. Although various studies using silencing approaches have supported the antiviral activity of the exo-siRNA pathway in mosquitoes, and despite strong similarities between the siRNA pathway in the Drosophila melanogaster model and mosquitoes, important questions remain unanswered. The antiviral activity of Ago2 against different arboviruses has been previously demonstrated. However, silencing of Ago2 had no effect on ZIKV replication, whereas Dcr2 knockout enhanced its replication. These findings raise the question as to the role of Ago2 and Dcr2 in the control of arboviruses from different viral families in mosquitoes. Using a newly established Ago2 knockout cell line, alongside the previously reported Dcr2 knockout cell line, we investigated the impact these proteins have on the modulation of different arboviral infections. Infection of Ago2 knockout cell line with alpha- and bunyaviruses resulted in an increase of viral replication, but not in the case of ZIKV. Analysis of small RNA sequencing data in the Ago2 knockout cells revealed a lack of methylated siRNAs from different sources, such as acute and persistently infecting viruses-, TE- and transcriptome-derived RNAs. The results confirmed the importance of the exo-siRNA pathway in the defense against arboviruses, but highlights variability in its response to different viruses and the impact the siRNA pathway proteins have in controlling viral replication. Moreover, this established Ago2 knockout cell line can be used for functional Ago2 studies, as well as research on the interplay between the RNAi pathways.


Subject(s)
Aedes/genetics , Aedes/virology , Arbovirus Infections/transmission , Arbovirus Infections/virology , Arboviruses/physiology , Argonaute Proteins/deficiency , Mosquito Vectors/genetics , Mosquito Vectors/virology , Animals , Cell Line , Gene Knockout Techniques , Host-Pathogen Interactions , RNA Interference , Virus Replication
2.
RNA ; 27(9): 991-1003, 2021 09.
Article in English | MEDLINE | ID: mdl-34108230

ABSTRACT

Mammalian RNA interference (RNAi) is often linked to the regulation of gene expression in the cytoplasm. Synthetic RNAs, however, can also act through the RNAi pathway to regulate transcription and splicing. While nuclear regulation by synthetic RNAs can be robust, a critical unanswered question is whether endogenous functions for nuclear RNAi exist in mammalian cells. Using enhanced crosslinking immunoprecipitation (eCLIP) in combination with RNA sequencing (RNA-seq) and multiple AGO knockout cell lines, we mapped AGO2 protein binding sites within nuclear RNA. The strongest AGO2 binding sites were mapped to micro RNAs (miRNAs). The most abundant miRNAs were distributed similarly between the cytoplasm and nucleus, providing no evidence for mechanisms that facilitate localization of miRNAs in one compartment versus the other. Beyond miRNAs, most statistically significant AGO2 binding was within introns. Splicing changes were confirmed by RT-PCR and recapitulated by synthetic miRNA mimics complementary to the sites of AGO2 binding. These data support the hypothesis that miRNAs can control gene splicing. While nuclear RNAi proteins have the potential to be natural regulatory mechanisms, careful study will be necessary to identify critical RNA drivers of normal physiology and disease.


Subject(s)
Alternative Splicing , Argonaute Proteins/genetics , Eukaryotic Initiation Factors/genetics , MicroRNAs/genetics , RNA, Nuclear/genetics , Argonaute Proteins/deficiency , Base Pairing , Base Sequence , Binding Sites , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Eukaryotic Initiation Factors/deficiency , Exons , HCT116 Cells , Humans , Immunoprecipitation , Introns , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Protein Binding , RNA, Nuclear/metabolism , Sequence Analysis, RNA
3.
RNA ; 27(9): 1004-1016, 2021 09.
Article in English | MEDLINE | ID: mdl-34108231

ABSTRACT

TNRC6 is a scaffolding protein that bridges interactions between small RNAs, argonaute (AGO) protein, and effector proteins to control gene expression. There are three paralogs in mammalian cells, TNRC6A, TNRC6B, and TNRC6C These paralogs have ∼40% amino acid sequence identity and the extent of their unique or redundant functions is unclear. Here, we use knockout cell lines, enhanced crosslinking immunoprecipitation (eCLIP), and high-throughput RNA sequencing (RNA-seq) to explore the roles of TNRC6 paralogs in RNA-mediated control of gene expression. We find that the paralogs are largely functionally redundant and changes in levels of gene expression are well-correlated with those observed in AGO knockout cell lines. Splicing changes observed in AGO knockout cell lines are also observed in TNRC6 knockout cells. These data further define the roles of the TNRC6 isoforms as part of the RNA interference (RNAi) machinery.


Subject(s)
Alternative Splicing , Autoantigens/genetics , RNA-Binding Proteins/genetics , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Autoantigens/metabolism , Binding Sites , Cell Line, Tumor , Exons , Gene Knockout Techniques , HCT116 Cells , Humans , Immunoprecipitation , Introns , Protein Binding , RNA-Binding Proteins/metabolism , Sequence Analysis, RNA
4.
Biochim Biophys Acta Gene Regul Mech ; 1864(6-7): 194704, 2021.
Article in English | MEDLINE | ID: mdl-33895310

ABSTRACT

BACKGROUND: RNA interference (RNAi) is a cellular mechanism used to fight various threats, including transposons, aberrant RNAs, and some types of viruses. This mechanism relies on the detection of dsRNA molecules, which through a pathway involving Dicer-2 (Dcr-2) and Argonaute 2 (AGO2), produces small interfering RNAs (siRNAs) that bind to the complementary RNAs triggering their degradation. METHODS: Using the cockroach Blattella germanica as a model, we examined AGO2 activity by depleting its mRNA using RNAi and analyzing the phenotypes produced. RESULTS: Depleting AGO2 expression had no remarkable effect on nymphal development or reproduction. dsRNA treatment triggered an immediate and transitory increase in AGO2 expression, independently of Dcr-2 action. In addition, we analyzed the siRNAs generated after injecting a heterologous dsRNA in control and AGO2-depleted animals. The results revealed that obtained siRNAs mapped non-uniformly along the dsRNA sequence. In AGO2-depleted animals, the proportion of 22 nucleotide reads was higher and accumulations of reads appeared in areas less well-represented in the controls. We also detected a preference for cytosine as the first nucleotide in controls that was significantly attenuated in AGO2-depleted individuals. CONCLUSIONS/GENERAL SIGNIFICANCE: The siRNAs produced from a dsRNA mapped heterogeneously along the length of the dsRNA and this arrangement depends on the dsRNA sequence. AGO2 exerts its role as nuclease on the siRNA duplexes independently of its action on the corresponding mRNA. This study sheds light on an extremely useful process for reverse genetics in laboratories, in addition to the design of more effective, specific, and eco-friendly pest-control strategies.


Subject(s)
Animals, Genetically Modified , Argonaute Proteins/deficiency , Blattellidae , Gene Silencing , Insect Proteins/deficiency , RNA, Small Interfering , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Argonaute Proteins/genetics , Blattellidae/genetics , Blattellidae/metabolism , Insect Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
5.
Endocrinology ; 162(4)2021 04 01.
Article in English | MEDLINE | ID: mdl-33567453

ABSTRACT

Argonaute 2 (Ago2) is the main component of the RNA-induced silencing complex. We recently showed that liver-specific Ago2-deficiency in mice (L-Ago2 knockout [KO] mice) enhances mitochondrial oxidation and alleviates obesity-associated pathophysiology. However, the precise mechanisms behind the role of hepatic Ago2 in regulating the mitochondrial oxidation associated with glucose metabolism are still unclear. Here, we show that hepatic Ago2 regulates the function of peroxisome proliferator-activated receptor α (PPARα) for oxidative metabolism. In both genetically and diet-induced severe obese conditions, L-Ago2 KO mice developed obesity and hepatic steatosis but exhibited improved glucose metabolism accompanied by lowered expression levels of pathologic microRNAs (miRNAs), including miR-802, miR-103/107, and miR-152, and enhanced expression of PPARα and its target genes regulating oxidative metabolism in the liver. We then investigated the role of hepatic Ago2 in the outcomes of vertical sleeve gastrectomy (VSG) in which PPARα plays a crucial role in a drastic transcription reprogram associated with improved glycemia post VSG. Whereas VSG reduced body weight and improved fatty liver in wild-type mice, these effects were not observed in hepatic Ago2-deficient mice. Conversely, glucose metabolism was improved in a hepatic Ago2-dependent manner post VSG. Treating Ago2-deficient primary hepatocytes with WY-14643, a PPARα agonist, showed that Ago2-deficiency enhances sensitivity to WY-14643 and increases expression of PPARα target genes and mitochondrial oxidation. Our findings suggest that hepatic Ago2 function is intrinsically associated with PPARα that links Ago2-mediated RNA silencing with mitochondrial functions for oxidation and obesity-associated pathophysiology.


Subject(s)
Argonaute Proteins/deficiency , Liver/metabolism , Obesity/metabolism , Obesity/surgery , PPAR alpha/metabolism , Animals , Argonaute Proteins/genetics , Bariatric Surgery , Glucose/metabolism , Glucose Tolerance Test , Glycemic Control , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/drug therapy , Obesity/genetics , Oxidative Stress , PPAR alpha/genetics , Pyrimidines/administration & dosage
6.
Nat Cell Biol ; 22(2): 235-245, 2020 02.
Article in English | MEDLINE | ID: mdl-32015436

ABSTRACT

PIWI-interacting RNAs (piRNAs) promote fertility in many animals. However, whether this is due to their conserved role in repressing repetitive elements (REs) remains unclear. Here, we show that the progressive loss of fertility in Caenorhabditis elegans lacking piRNAs is not caused by derepression of REs or other piRNA targets but, rather, is mediated by epigenetic silencing of all of the replicative histone genes. In the absence of piRNAs, downstream components of the piRNA pathway relocalize from germ granules and piRNA targets to histone mRNAs to synthesize antisense small RNAs (sRNAs) and induce transgenerational silencing. Removal of the downstream components of the piRNA pathway restores histone mRNA expression and fertility in piRNA mutants, and the inheritance of histone sRNAs in wild-type worms adversely affects their fertility for multiple generations. We conclude that sRNA-mediated silencing of histone genes impairs the fertility of piRNA mutants and may serve to maintain piRNAs across evolution.


Subject(s)
Argonaute Proteins/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Gene Silencing , Histones/genetics , RNA, Small Interfering/genetics , Animals , Animals, Genetically Modified , Argonaute Proteins/deficiency , Argonaute Proteins/metabolism , Biological Evolution , CRISPR-Cas Systems , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Fertility/genetics , Gene Editing , Histones/metabolism , Inheritance Patterns , Mutation , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Repetitive Sequences, Nucleic Acid
7.
Stem Cells ; 37(6): 803-812, 2019 06.
Article in English | MEDLINE | ID: mdl-30805989

ABSTRACT

Remodeling of the gene regulatory network in cells is believed to be a prerequisite for their lineage reprogramming. However, its key regulatory factors are not yet elucidated. In this article, we investigate the role of PIWI proteins and provide evidence that one of them, MIWI2, is elicited during transdifferentiation of fibroblasts into hepatocyte-like cells. In coincidence with the peak expression of MIWI2, we identified the appearance of a unique intermediate epigenetic state characterized by a specific Piwi-interacting RNA (piRNA) profile consisting of 219 novel sequences. Knockout of MIWI2 greatly improved the formation of the induced hepatocytes, whereas overexpression of exogenous MIWI2 completely abolished the stimulated effect. A bioinformatics analysis of piRNA interaction network, followed by experimental validation, revealed the Notch signaling pathway as one of the immediate effectors of MIWI2. Altogether, our results show for the first time that temporal expression of MIWI2 contributes negatively to cell plasticity not only in germline, but also in developed cells, such as mouse fibroblasts. Stem Cells 2019;37:803-812.


Subject(s)
Argonaute Proteins/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Hepatocytes/metabolism , RNA, Small Interfering/genetics , Albumins/genetics , Albumins/metabolism , Animals , Argonaute Proteins/deficiency , CRISPR-Cas Systems , Cell Lineage/genetics , Cell Transdifferentiation/genetics , Fibroblasts/cytology , Gene Regulatory Networks , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/metabolism , Hepatocytes/cytology , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Knockout , RNA, Small Interfering/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Transduction, Genetic
8.
J Med Chem ; 61(3): 734-744, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29376650

ABSTRACT

(E)-Vinylphosphonate ((E)-VP), a metabolically stable phosphate mimic at the 5'-end of the antisense strand, enhances the in vivo potency of siRNA. Here we describe a straightforward synthetic approach to incorporate a nucleotide carrying a vinylphosphonate (VP) moiety at the 5'-end of oligonucleotides under standard solid-phase synthesis and deprotection conditions by utilizing pivaloyloxymethyl (POM) protected VP-nucleoside phosphoramidites. The POM protection enhances scope and scalability of 5'-VP-modified oligonucleotides and, in a broader sense, the synthesis of oligonucleotides modified with phosphonate moieties. Trivalent N-acetylgalactosamine-conjugated small interfering RNA (GalNAc-siRNA) comprising (E)-geometrical isomer of VP showed improved RISC loading with robust RNAi-mediated gene silencing in mice compared to the corresponding (Z)-isomer despite similar tissue accumulation. We also obtained structural insights into why bulkier 2'-ribosugar substitutions such as 2'-O-[2-(methylamino)-2-oxoethyl] are well tolerated only when combined with 5'-(E)-VP.


Subject(s)
Organophosphonates/chemistry , Organophosphonates/chemical synthesis , RNA, Small Interfering/chemistry , Animals , Argonaute Proteins/chemistry , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Base Sequence , Chemistry Techniques, Synthetic , Gene Silencing , Mice , Models, Molecular , Protein Domains , RNA, Small Interfering/genetics , Stereoisomerism
9.
Reproduction ; 154(6): 723-733, 2017 12.
Article in English | MEDLINE | ID: mdl-28871057

ABSTRACT

The chromatoid body (CB) is a specific cloud-like structure in the cytoplasm of haploid spermatids. Recent findings indicate that CB is identified as a male germ cell-specific RNA storage and processing center, but its function has remained elusive for decades. In somatic cells, KH-type splicing regulatory protein (KSRP) is involved in regulating gene expression and maturation of select microRNAs (miRNAs). However, the function of KSRP in spermatogenesis remains unclear. In this study, we showed that KSRP partly localizes in CB, as a component of CB. KSRP interacts with proteins (mouse VASA homolog (MVH), polyadenylate-binding protein 1 (PABP1) and polyadenylate-binding protein 2 (PABP2)), mRNAs (Tnp2 and Odf1) and microRNAs (microRNA-182) in mouse CB. Moreover, KSRP may regulate the integrity of CB via DDX5-miRNA-182 pathway. In addition, we found abnormal expressions of CB component in testes of Ksrp-knockout mice and of patients with hypospermatogenesis. Thus, our results provide mechanistic insight into the role of KSRP in spermatogenesis.


Subject(s)
RNA-Binding Proteins/metabolism , Spermatids/metabolism , Spermatogenesis , Trans-Activators/metabolism , Adult , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Case-Control Studies , Cells, Cultured , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligospermia/genetics , Oligospermia/metabolism , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Protein II/genetics , Poly(A)-Binding Protein II/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Trans-Activators/deficiency , Trans-Activators/genetics , Young Adult
10.
Sci Rep ; 7(1): 5462, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28710366

ABSTRACT

The unicellular alga Chlamydomonas reinhardtii contains many types of small RNAs (sRNAs) but the biological role(s) of bona fide microRNAs (miRNAs) remains unclear. To address their possible function(s) in responses to nutrient availability, we examined miRNA expression in cells cultured under different trophic conditions (mixotrophic in the presence of acetate or photoautotrophic in the presence or absence of nitrogen). We also reanalyzed miRNA expression data in Chlamydomonas subject to sulfur or phosphate deprivation. Several miRNAs were differentially expressed under the various trophic conditions. However, in transcriptome analyses, the majority of their predicted targets did not show expected changes in transcript abundance, suggesting that they are not subject to miRNA-mediated RNA degradation. Mutant strains, defective in sRNAs or in ARGONAUTE3 (a key component of sRNA-mediated gene silencing), did not display major phenotypic defects when grown under multiple nutritional regimes. Additionally, Chlamydomonas miRNAs were not conserved, even in algae of the closely related Volvocaceae family, and many showed features resembling those of recently evolved, species-specific miRNAs in the genus Arabidopsis. Our results suggest that, in C. reinhardtii, miRNAs might be subject to relatively fast evolution and have only a minor, largely modulatory role in gene regulation under diverse trophic states.


Subject(s)
Chlamydomonas reinhardtii/genetics , Gene Expression Regulation, Plant , MicroRNAs/genetics , Phosphates/deficiency , RNA, Algal/genetics , Sulfur/metabolism , Acetic Acid/metabolism , Acetic Acid/pharmacology , Arabidopsis/genetics , Arabidopsis/metabolism , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Biological Evolution , Chlamydomonas reinhardtii/drug effects , Chlamydomonas reinhardtii/metabolism , MicroRNAs/metabolism , Nitrogen/metabolism , Nitrogen/pharmacology , Phosphates/pharmacology , Phylogeny , RNA, Algal/metabolism , Sulfur/pharmacology
11.
Nat Struct Mol Biol ; 24(7): 604-606, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530707

ABSTRACT

In mice, the pathway involving PIWI and PIWI-interacting RNA (PIWI-piRNA) is essential to re-establish transposon silencing during male-germline reprogramming. The cytoplasmic PIWI protein MILI mediates piRNA-guided transposon RNA cleavage as well as piRNA amplification. MIWI2's binding to piRNA and its nuclear localization are proposed to be dependent upon MILI function. Here, we demonstrate the existence of a piRNA biogenesis pathway that sustains partial MIWI2 function and reprogramming activity in the absence of MILI.


Subject(s)
Argonaute Proteins/metabolism , Gene Expression Regulation , RNA, Small Interfering/metabolism , Animals , Argonaute Proteins/deficiency , Mice , Mice, Knockout
12.
J Chem Inf Model ; 57(4): 883-896, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28287733

ABSTRACT

The use of chemical modifications in small interfering RNAs (siRNAs) is warranted to impart drug-like properties. However, certain chemical modifications especially those on the sugar have deleterious effects on the RNA interference (RNAi) when they are placed at key positions in the seed region of an siRNA guide strand. In order to probe the effect of chemically modified siRNAs [(2'-O-methyl, 4'-C-aminomethyl-2'-O-methyl, 2'-O-(2-methoxyethyl), and 2'-O-benzyl] on human Argonaute 2 (hAGO2), the catalytic engine of RNAi, we have developed a model of its open conformation. Results from microsecond MD simulations of 15 different siRNA-hAGO2 complexes provide insights about how the key noncovalent interactions and conformational changes at the seed region are modulated, depending upon the nature and position of chemical modifications. Such modification induced structural changes can affect siRNA loading into hAGO2, which may influence RNAi activity. Our studies show that microsecond MD simulations can provide useful information for the design of therapeutically relevant siRNAs.


Subject(s)
Argonaute Proteins/metabolism , Molecular Dynamics Simulation , RNA, Small Interfering/metabolism , Argonaute Proteins/chemistry , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Base Pairing , Base Sequence , Binding Sites , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Phosphates/metabolism , Protein Binding , Protein Conformation , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , Thermodynamics
13.
J Virol ; 91(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28331090

ABSTRACT

P-element-induced wimpy-like (Piwil) proteins restrict the replication of mobile genetic elements in the germ line. They are also expressed in many transformed cell lines. In this study, we discovered that the human Piwil 2 (Hili) protein can also inhibit HIV replication, especially in activated CD4+ T cells that are the preferred target cells for this virus in the infected host. Although resting cells did not express Hili, its expression was rapidly induced following T cell activation. In these cells and transformed cell lines, depletion of Hili increased levels of viral proteins and new viral particles. Further studies revealed that Hili binds to tRNA. Some of the tRNAs represent rare tRNA species, whose codons are overrepresented in the viral genome. Targeting tRNAArg(UCU) with an antisense oligonucleotide replicated effects of Hili and also inhibited HIV replication. Finally, Hili also inhibited the retrotransposition of the endogenous intracysternal A particle (IAP) by a similar mechanism. Thus, Hili joins a list of host proteins that inhibit the replication of HIV and other mobile genetic elements.IMPORTANCE Piwil proteins inhibit the movement of mobile genetic elements in the germ line. In their absence, sperm does not form and male mice are sterile. This inhibition is thought to occur via small Piwi-interacting RNAs (piRNAs). However, in some species and in human somatic cells, Piwil proteins bind primarily to tRNA. In this report, we demonstrate that human Piwil proteins, especially Hili, not only bind to select tRNA species, including rare tRNAs, but also inhibit HIV replication. Importantly, T cell activation induces the expression of Hili in CD4+ T cells. Since Hili also inhibited the movement of an endogenous retrovirus (IAP), our finding shed new light on this intracellular resistance to exogenous and endogenous retroviruses as well as other mobile genetic elements.


Subject(s)
Argonaute Proteins/metabolism , HIV-1/physiology , Lymphocyte Activation , Virus Replication , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Argonaute Proteins/immunology , Cell Line , Endogenous Retroviruses/metabolism , HEK293 Cells , HIV-1/genetics , HeLa Cells , Humans , Oligonucleotides, Antisense/genetics , Protein Binding , RNA, Small Interfering/metabolism , RNA, Transfer/metabolism , RNA, Transfer, Arg/genetics , RNA, Transfer, Arg/metabolism , T-Lymphocytes/virology
14.
J Am Chem Soc ; 138(28): 8694-7, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27380263

ABSTRACT

Incorporation of chemical modifications into small interfering RNAs (siRNAs) increases their metabolic stability and improves their tissue distribution. However, how these modifications impact interactions with Argonaute-2 (Ago2), the molecular target of siRNAs, is not known. Herein we present the crystal structure of human Ago2 bound to a metabolically stable siRNA containing extensive backbone modifications. Comparison to the structure of an equivalent unmodified-siRNA complex indicates that the structure of Ago2 is relatively unaffected by chemical modifications in the bound siRNA. In contrast, the modified siRNA appears to be much more plastic and shifts, relative to the unmodified siRNA, to optimize contacts with Ago2. Structure-activity analysis reveals that even major conformational perturbations in the 3' half of the siRNA seed region have a relatively modest effect on knockdown potency. These findings provide an explanation for a variety of modification patterns tolerated in siRNAs and a structural basis for advancing therapeutic siRNA design.


Subject(s)
Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Crystallography, X-Ray , Humans , Protein Binding , RNA Interference
15.
Stem Cells ; 34(5): 1343-53, 2016 05.
Article in English | MEDLINE | ID: mdl-26850790

ABSTRACT

Hematopoietic stem cells (HSCs) are capable of both self-renewing throughout the lifetime of an organism and differentiating into all lineages of the blood system. A proper balance between quiescence and proliferation is critical for the self-renewal and functions of HSCs. The choice of HSCs to remain quiescent or to enter proliferation has been tightly regulated by a variety of cell intrinsic and extrinsic pathways. Identifying molecular players that control HSC quiescence and proliferation may lead to new treatment strategies and therapeutic interventions for hematologic disorders. To identify the functions of the slicer endonuclease Argonaute (Ago) 2 in the physiology of HSCs, we generated Ago2(Hem-KO) mice, that are deficient for Ago2 in HSCs and in their progeny. Analysis of Ago2(Hem-KO) mice indicated that a loss of Ago2 results in reduced HSC pool size and altered frequencies of hematopoietic progenitors. Ago2 deficient HSCs exhibit defective multilineage differentiation capacities and diminished repopulation abilities, in a cell intrinsic manner. Interestingly, Ago2 mutant HSCs remain largely quiescent and show reduced entry into cell cycle. Genome-wide transcriptome studies and gene set enrichment analysis revealed that Ago2 deficient HSCs downregulate the "HSC signature" and upregulate the "lineage signature." Moreover, our analysis on transcription factors (TFs) identified that a loss of Ago2 is sufficient to alter the "molecular signature" and "TF networks" that control the quiescent and proliferative states of HSCs. In essence, our study identified Ago2 as a key determinant of quiescence exit in HSCs. Stem Cells 2016;34:1343-1353.


Subject(s)
Argonaute Proteins/metabolism , Cell Cycle , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Argonaute Proteins/deficiency , Blood Cell Count , Body Weight , Bone Marrow/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Gene Regulatory Networks , Mice, Knockout , Mutation/genetics , Transcription Factors/metabolism , Transcriptome
16.
BMC Cancer ; 15: 811, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26506848

ABSTRACT

BACKGROUND: Stem cell protein Piwil1 functions as an oncogene in various tumor types. However, the exact function and mechanism of Piwil1 in endometrial cancer remains unclear. METHODS: The expression of Piwil1 and its relationships with clinicopathological factors were investigated using immunohistochemistry. Up- or down-regulation of Piwil1 were achieved by stable or transient transfection with plasmids or short hairpin RNA (shRNA). Effects of Piwil1 on cancer cells viability, invasion and migration were evaluated by MTT, plate colony formation, transwell assay and nude mouse tumor xenograft assay. The stem-like properties of endometrial cancer cells was detected by spheroid formation assay. Effects of Piwil1 on expression levels of target genes were detected by qRT-PCR, western blotting and Immunofluorescence. RESULTS: Compared with atypical hyperplasia and normal tissues, Piwil1 was much higher in endometrial carcinoma tissues. We found that Piwil1 expression was significantly correlated with FIGO stage, lymphovascular space involvement, lymph node metastasis and level of myometrial invasion. Overexpression of Piwil1 functioned to maintain stem-like characteristics, including enhancing tumor cell viability, migration, invasion and sphere-forming activity. Conversely, Piwil1 knockdown inhibited cell viability, migration, invasion, sphere-forming activity in vitro and tumor formation in xenograft model in vivo. Furthermore, study of the expression of epithelial and mesenchymal markers showed that Piwil1 was responsible for an EMT-like phenotype associated with an increase in mesenchymal markers and suppression of E-cadherin. Moreover, Piwil1 augmented expression levels of CD44 and ALDH1 expression, two known endometrial CSC markers, as well as other stemness-associated genes. CONCLUSIONS: Our results suggested that stem cell protein Piwil1 play important roles in regulating EMT and the acquisition of stem-like properties of endometrial cancer cells. Therefore, it indicated that Piwil1 may represent a promising target for developing a novel treatment strategy for endometrial cancer.


Subject(s)
Argonaute Proteins/biosynthesis , Endometrial Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Neoplastic Stem Cells/metabolism , Animals , Argonaute Proteins/deficiency , Cell Line, Tumor , Endometrial Neoplasms/pathology , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays/methods
17.
PLoS One ; 10(10): e0140291, 2015.
Article in English | MEDLINE | ID: mdl-26451954

ABSTRACT

EV71 (enterovirus 71) RNA contains an internal ribosomal entry site (IRES) that directs cap-independent initiation of translation. IRES-dependent translation requires the host's translation initiation factors and IRES-associated trans-acting factors (ITAFs). We reported recently that mRNA decay factor AUF1 is a negative-acting ITAF that binds IRES stem-loop II. We also reported that the small RNA-processing enzyme Dicer produces at least four small RNAs (vsRNAs) from the EV71 IRES. One of these, vsRNA1, derived from IRES stem-loop II, reduces IRES activity and virus replication. Since its mechanism of action is unknown, we hypothesized that it might control association of ITAFs with the IRES. Here, we identified the mRNA stability factor HuR and the RISC subunit Argonaute 2 (Ago2) as two ITAFs that bind stem-loop II. In contrast to AUF1, HuR and Ago2 promote EV71 IRES activity and virus replication. In vitro RNA-binding assays revealed that vsRNA1 can alter association of Ago2, HuR, and AUF1 with stem-loop II. This presents a possible mechanism by which vsRNA1 could control viral translation and replication.


Subject(s)
Argonaute Proteins/metabolism , ELAV-Like Protein 1/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/physiology , Internal Ribosome Entry Sites , Protein Biosynthesis , Virus Replication/genetics , 5' Untranslated Regions/genetics , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Cell Line, Tumor , Chlorocebus aethiops , ELAV-Like Protein 1/deficiency , ELAV-Like Protein 1/genetics , Enterovirus A, Human/metabolism , Gene Knockdown Techniques , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Inverted Repeat Sequences/genetics , Protein Binding , RNA, Viral/genetics , RNA, Viral/metabolism , Vero Cells
18.
Biochem Biophys Res Commun ; 464(4): 1066-1071, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26188511

ABSTRACT

Post-translational modification of substrate proteins by small ubiquitin-like modifier (SUMO) regulates a vast array of cellular processes. SUMOylation occurs through three sequential enzymatic steps termed E1, E2 and E3. Substrate selection can be determined through interactions between the target protein and the SUMO E2 conjugating enzyme Ubc9 and specificity can be enhanced by substrate interactions with E3 ligase enzymes. We used the putative substrate recognition (PINIT) domain from the SUMO E3 PIAS3 as bait to identify potential SUMO substrates. One protein identified was Argonaute-2 (Ago2), which mediates RNA-induced gene silencing through binding small RNAs and promoting degradation of complimentary target mRNAs. We show that Ago2 can be SUMOylated in mammalian cells by both SUMO1 and SUMO2. SUMOylation occurs primarily at K402, and mutation of the SUMO consensus site surrounding this lysine reduces Ago2-mediated siRNA-induced silencing in a luciferase-based reporter assay. These results identify SUMOylation as a potential regulator of Ago2 activity and open new avenues for research into the mechanisms underlying the regulation of RNA-induced gene silencing.


Subject(s)
Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , RNA Interference , Sumoylation , Amino Acid Sequence , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Binding Sites/genetics , Cells, Cultured , Consensus Sequence , Gene Knockout Techniques , Humans , Mice , Mutagenesis, Site-Directed , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Substrate Specificity
19.
BMC Genomics ; 15: 1157, 2014 Dec 22.
Article in English | MEDLINE | ID: mdl-25534009

ABSTRACT

BACKGROUND: Small RNA-guided transcriptional silencing (nuclear RNAi) is fundamental to genome integrity and epigenetic inheritance. Despite recent progress in identifying the capability and genetic requirements for nuclear RNAi in Caenorhabditis elegans, the natural targets and cellular functions of nuclear RNAi remain elusive. METHODS: To resolve this gap, we coordinately examined the genome-wide profiles of transcription, histone H3 lysine 9 methylation (H3K9me) and endogenous siRNAs of a germline nuclear Argonaute (hrde-1/wago-9) mutant and identified regions on which transcription activity is markedly increased and/or H3K9me level is markedly decreased relative to wild type animals. RESULTS: Our data revealed a distinct set of native targets of germline nuclear RNAi, with the H3K9me response exhibiting both overlapping and non-overlapping distribution with the transcriptional silencing response. Interestingly LTR retrotransposons, but not DNA transposons, are highly enriched in the targets of germline nuclear RNAi. The genomic distribution of the native targets is highly constrained, with >99% of the identified targets present in five autosomes but not in the sex chromosome. By contrast, HRDE-1-associated small RNAs correspond to all chromosomes. In addition, we found that the piRNA pathway is not required for germline nuclear RNAi activity on native targets. CONCLUSION: Germline nuclear RNAi in C. elegans is required to silence retrotransposons but not DNA transposon. Transcriptional silencing and H3K9me can occur independently of each other on the native targets of nuclear RNAi in C. elegans. Our results rule out a simple model in which nuclear Argonaute protein-associated-small RNAs are sufficient to trigger germline nuclear RNAi responses. In addition, the piRNA pathway and germline nuclear RNAi are specialized to target different types of foreign genetic elements for genome surveillance in C. elegans.


Subject(s)
Caenorhabditis elegans/genetics , Histones/chemistry , Histones/metabolism , RNA Interference , RNA, Small Interfering/genetics , Transcription, Genetic/genetics , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Caenorhabditis elegans/cytology , Chromatin/genetics , Gene Expression Profiling , Lysine/metabolism , Methylation , RNA Polymerase II/metabolism , Retroelements/genetics
20.
PLoS One ; 9(5): e94538, 2014.
Article in English | MEDLINE | ID: mdl-24788663

ABSTRACT

Gene silencing and RNA interference are major cellular processes that control gene expression via the cleavage of target mRNA. Eukaryotic translation initiation factor 2C2 (EIF2C2, Argonaute protein 2, Ago2) is considered to be the major player of RNAi as it is the core component of RISC complexes. While a considerable amount of research has focused on RNA interference and its associated mechanisms, the nature and mechanisms of nucleotide recognition by the PAZ domain of EIF2C2/Ago2 have not yet been characterized. Here, we demonstrate that the EIF2C2/Ago2 PAZ domain has an inherent lack of binding to adenine nucleotides, a feature that highlights the poor binding of 3'-adenylated RNAs with the PAZ domain as well as the selective high trimming of the 3'-ends of miRNA containing adenine nucleotides. We further show that the PAZ domain selectively binds all ribonucleotides (except adenosine), whereas it poorly recognizes deoxyribonucleotides. In this context, the modification of dTMP to its ribonucleotide analogue gave a drastic improvement of binding enthalpy and, hence, binding affinity. Additionally, higher in vivo gene silencing efficacy was correlated with the stronger PAZ domain binders. These findings provide new insights into the nature of the interactions of the EIF2C2/Ago2 PAZ domain.


Subject(s)
Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Energy Metabolism , Nucleotides/metabolism , RNA Interference , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Base Sequence , Entropy , HeLa Cells , Hot Temperature , Humans , Models, Molecular , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL