Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Mol Cell ; 65(3): 490-503.e7, 2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28132840

ABSTRACT

Environmental cues provoke rapid transitions in gene expression to support growth and cellular plasticity through incompletely understood mechanisms. Lin28 RNA-binding proteins have evolutionarily conserved roles in post-transcriptional coordination of pro-growth gene expression, but signaling pathways allowing trophic stimuli to induce Lin28 have remained uncharacterized. We find that Lin28a protein exhibits rapid basal turnover in neurons and that mitogen-activated protein kinase (MAPK)-dependent phosphorylation of the RNA-silencing factor HIV TAR-RNA-binding protein (TRBP) promotes binding and stabilization of Lin28a, but not Lin28b, with an accompanying reduction in Lin28-regulated miRNAs, downstream of brain-derived neurotrophic factor (BDNF). Binding of Lin28a to TRBP in vitro is also enhanced by phospho-mimic TRBP. Further, phospho-TRBP recapitulates BDNF-induced neuronal dendritic spine growth in a Lin28a-dependent manner. Finally, we demonstrate MAPK-dependent TRBP and Lin28a induction, with physiological function in growth and survival, downstream of diverse growth factors in multiple primary cell types, supporting a broad role for this pathway in trophic responses.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dendritic Spines/metabolism , Mitogen-Activated Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Proliferation , Cell Survival , HEK293 Cells , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Humans , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Mice , Neurons/metabolism , Phosphorylation
2.
Cell Mol Life Sci ; 81(1): 53, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38261114

ABSTRACT

The human let-7 miRNA family consists of thirteen members that play critical roles in many biological processes, including development timing and tumor suppression, and their levels are disrupted in several diseases. Dicer is the endoribonuclease responsible for processing the precursor miRNA (pre-miRNA) to yield the mature miRNA, and thereby plays a crucial role in controlling the cellular levels of let-7 miRNAs. It is well established that the sequence and structural features of pre-miRNA hairpins such as the 5'-phosphate, the apical loop, and the 2-nt 3'-overhang are important for the processing activity of Dicer. Exceptionally, nine precursors of the let-7 family (pre-let-7) contain a 1-nt 3'-overhang and get mono-uridylated in vivo, presumably to allow efficient processing by Dicer. Pre-let-7 are also oligo-uridylated in vivo to promote their degradation and likely prevent their efficient processing by Dicer. In this study, we systematically investigated the impact of sequence and structural features of all human let-7 pre-miRNAs, including their 3'-end modifications, on Dicer binding and processing. Through the combination of SHAPE structural probing, in vitro binding and kinetic studies using purified human Dicer, we show that despite structural discrepancies among pre-let-7 RNAs, Dicer exhibits remarkable promiscuity in binding and cleaving these substrates. Moreover, the 1- or 2-nt 3'-overhang, 3'-mono-uridylation, and 3'-oligo-uridylation of pre-let-7 substrates appear to have little effect on Dicer binding and cleavage rates. Thus, this study extends current knowledge regarding the broad substrate specificity of Dicer and provides novel insight regarding the effect of 3'-modifications on binding and cleavage by Dicer.


Subject(s)
DEAD-box RNA Helicases , MicroRNAs , Ribonuclease III , Humans , Kinetics , MicroRNAs/genetics , Phosphates , Substrate Specificity , DEAD-box RNA Helicases/genetics , Ribonuclease III/genetics
3.
Biochem Biophys Res Commun ; 658: 122-127, 2023 05 28.
Article in English | MEDLINE | ID: mdl-37030066

ABSTRACT

Viral infection induces diverse cellular immune responses. Some viruses induce the production of antiviral cytokines, alterations of endogenous gene expression, and apoptosis; however, other viruses replicate without inducing such responses, enabling them to persistently infect cells. Infection by Borna disease virus type 1 (BoDV-1) can result in fatal immune-mediated encephalitis, including in humans, yet infection of cells in vitro is generally persistent. The regulatory mechanisms underlying this persistent infection remain unclear. Here, we show that an enhancer of RNA-silencing, TRBP, positively regulates BoDV RNA level in human cells. Knockdown of TRBP decreased BoDV RNA levels in persistently-infected cells, whereas overexpression of TRBP increased BoDV RNA levels. To investigate the mechanism underlying this phenomenon, we performed immunoprecipitation assays and found that TRBP interacts with BoDV RNA. Furthermore, we performed cell fractionation, which revealed that persistent infection with BoDV does not alter the localization of TRBP and other RNA silencing factors in cells. Our results showed the regulation of persistent BoDV infection by RNA-silencing factors in human cells.


Subject(s)
Borna Disease , Borna disease virus , Animals , Humans , Borna disease virus/genetics , Borna Disease/genetics , Borna Disease/metabolism , RNA Interference , Persistent Infection , RNA
4.
EMBO J ; 37(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29449323

ABSTRACT

The accurate cleavage of pre-micro(mi)RNAs by Dicer and mi/siRNA guide strand selection are important steps in forming the RNA-induced silencing complex (RISC). The role of Dicer binding partner TRBP in these processes remains poorly understood. Here, we solved the solution structure of the two N-terminal dsRNA binding domains (dsRBDs) of TRBP in complex with a functionally asymmetric siRNA using NMR, EPR, and single-molecule spectroscopy. We find that siRNA recognition by the dsRBDs is not sequence-specific but rather depends on the RNA shape. The two dsRBDs can swap their binding sites, giving rise to two equally populated, pseudo-symmetrical complexes, showing that TRBP is not a primary sensor of siRNA asymmetry. Using our structure to model a Dicer-TRBP-siRNA ternary complex, we show that TRBP's dsRBDs and Dicer's RNase III domains bind a canonical 19 base pair siRNA on opposite sides, supporting a mechanism whereby TRBP influences Dicer-mediated cleavage accuracy by binding the dsRNA region of the pre-miRNA during Dicer cleavage.


Subject(s)
Double-Stranded RNA Binding Motif , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Models, Molecular , Spectrum Analysis/methods
5.
Biochem J ; 478(3): 493-510, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33459340

ABSTRACT

An integral aspect of innate immunity is the ability to detect foreign molecules of viral origin to initiate antiviral signaling via pattern recognition receptors (PRRs). One such receptor is the RNA helicase retinoic acid inducible gene 1 (RIG-I), which detects and is activated by 5'triphosphate uncapped double stranded RNA (dsRNA) as well as the cytoplasmic viral mimic dsRNA polyI:C. Once activated, RIG-I's CARD domains oligomerize and initiate downstream signaling via mitochondrial antiviral signaling protein (MAVS), ultimately inducing interferon (IFN) production. Another dsRNA binding protein PACT, originally identified as the cellular protein activator of dsRNA-activated protein kinase (PKR), is known to enhance RIG-I signaling in response to polyI:C treatment, in part by stimulating RIG-I's ATPase and helicase activities. TAR-RNA-binding protein (TRBP), which is ∼45% homologous to PACT, inhibits PKR signaling by binding to PKR as well as by sequestration of its' activators, dsRNA and PACT. Despite the extensive homology and similar structure of PACT and TRBP, the role of TRBP has not been explored much in RIG-I signaling. This work focuses on the effect of TRBP on RIG-I signaling and IFN production. Our results indicate that TRBP acts as an inhibitor of RIG-I signaling in a PACT- and PKR-independent manner. Surprisingly, this inhibition is independent of TRBP's post-translational modifications that are important for other signaling functions of TRBP, but TRBP's dsRNA-binding ability is essential. Our work has major implications on viral susceptibility, disease progression, and antiviral immunity as it demonstrates the regulatory interplay between PACT and TRBP IFN production.


Subject(s)
Carrier Proteins/physiology , DEAD Box Protein 58/physiology , RNA-Binding Proteins/physiology , Receptors, Immunologic/physiology , Signal Transduction/physiology , Active Transport, Cell Nucleus , Adenosine Triphosphate/metabolism , Animals , Fibroblasts , Genes, Reporter , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferons/physiology , Mice , Models, Biological , Mutation , Phosphorylation , Poly I-C/pharmacology , Protein Binding , Protein Domains , Protein Processing, Post-Translational , RNA, Double-Stranded/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
6.
Development ; 144(5): 778-783, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28174252

ABSTRACT

Transactivation response element RNA-binding protein (TRBP; TARBP2) is known to play important roles in human immunodeficiency virus (HIV) replication and microRNA biogenesis. However, recent studies implicate TRBP in a variety of biological processes as a mediator of cross-talk between signal transduction pathways. Here, we provide the first evidence that TRBP is required for efficient neurosphere formation and for the expression of neural stem cell markers and Notch target genes in primary neural progenitor cells in vitro Consistent with this, introduction of TRBP into the mouse embryonic brain in utero increased the fraction of cells expressing Sox2 in the ventricular zone. We also show that TRBP physically interacts with the Notch transcriptional coactivation complex through C promoter-binding factor 1 (CBF1; RBPJ) and strengthens the association between the Notch intracellular domain (NICD) and CBF1, resulting in increased NICD recruitment to the promoter region of a Notch target gene. Our data indicate that TRBP is a novel transcriptional coactivator of the Notch signaling pathway, playing an important role in neural stem cell regulation during mammalian brain development.


Subject(s)
Neural Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Receptors, Notch/metabolism , Transcriptional Activation , Animals , Brain/metabolism , Cell Nucleus/metabolism , Central Nervous System/embryology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , In Situ Hybridization , Mice , MicroRNAs/metabolism , Promoter Regions, Genetic , Signal Transduction
7.
Crit Rev Biochem Mol Biol ; 51(3): 121-34, 2016.
Article in English | MEDLINE | ID: mdl-26628006

ABSTRACT

MicroRNAs (miRNAs) are integral to the gene regulatory network. A single miRNA is capable of controlling the expression of hundreds of protein coding genes and modulate a wide spectrum of biological functions, such as proliferation, differentiation, stress responses, DNA repair, cell adhesion, motility, inflammation, cell survival, senescence and apoptosis, all of which are fundamental to tumorigenesis. Overexpression, genetic amplification, and gain-of-function mutation of oncogenic miRNAs ("onco-miRs") as well as genetic deletion and loss-of-function mutation of tumor suppressor miRNAs ("suppressor-miRs") are linked to human cancer. In addition to the dysregulation of a specific onco-miR or suppressor-miRs, changes in global miRNA levels resulting from a defective miRNA biogenesis pathway play a role in tumorigenesis. The function of individual onco-miRs and suppressor-miRs and their target genes in cancer has been described in many different articles elsewhere. In this review, we primarily focus on the recent development regarding the dysregulation of the miRNA biogenesis pathway and its contribution to cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/genetics , Animals , Argonaute Proteins/genetics , Gene Regulatory Networks , Humans , Protein Processing, Post-Translational , Ribonuclease III/genetics , Transcription, Genetic
8.
J Cell Biochem ; 119(4): 3598-3607, 2018 04.
Article in English | MEDLINE | ID: mdl-29231267

ABSTRACT

PACT is a stress-modulated activator of protein kinase PKR (protein kinase, RNA activated), which is involved in antiviral innate immune responses and stress-induced apoptosis. Stress-induced phosphorylation of PACT is essential for PACT's increased association with PKR leading to PKR activation, phosphorylation of translation initiation factor eIF2α, inhibition of protein synthesis, and apoptosis. PACT-induced PKR activation is negatively regulated by TRBP (transactivation response element RNA-binding protein), which dissociates from PACT after PACT phosphorylation in response to stress signals. The conserved double-stranded RNA binding motifs (dsRBMs) in PKR, PACT, and TRBP mediate protein-protein interactions, and the stress-dependent phosphorylation of PACT changes the relative strengths of PKR-PACT, PACT-TRBP, and PACT-PACT interactions to bring about a timely and transient PKR activation. This regulates the general kinetics as well as level of eIF2α phosphorylation, thereby influencing the cellular response to stress either as recovery and survival or elimination by apoptosis. In the present study, we evaluated the effect of specific mutations within PACT's two evolutionarily conserved dsRBMs on dsRNA-binding, and protein-protein interactions between PKR, PACT, and TRBP. Our data show that the two motifs contribute to varying extents in dsRNA binding, and protein interactions. These findings indicate that although the dsRBM motifs have high sequence conservation, their functional contribution in the context of the whole proteins needs to be determined by mutational analysis. Furthermore, using a PACT mutant that is deficient in PACT-PACT interaction but competent for PACT-PKR interaction, we demonstrate that PACT-PACT interaction is essential for efficient PKR activation.


Subject(s)
Double-Stranded RNA Binding Motif/physiology , RNA, Double-Stranded/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , COS Cells , Chlorocebus aethiops , Double-Stranded RNA Binding Motif/genetics , HeLa Cells , Humans , Phosphorylation/genetics , Phosphorylation/physiology , Protein Binding/genetics , Protein Binding/physiology , RNA, Double-Stranded/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Two-Hybrid System Techniques , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
9.
Methods ; 105: 109-18, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27012177

ABSTRACT

TRBP, one of double strand RNA binding proteins (dsRBPs), is an essential cofactor of Dicer in the RNA interference pathway. Previously we reported that TRBP exhibits repetitive diffusion activity on double strand (ds)RNA in an ATP independent manner. In the TRBP-Dicer complex, the diffusion mobility of TRBP facilitates Dicer-mediated RNA cleavage. Such repetitive diffusion of dsRBPs on a nucleic acid at the nanometer scale can be appropriately captured by several single molecule detection techniques. Here, we provide a step-by-step guide to four different single molecule fluorescence assays by which the diffusion activity of dsRBPs on dsRNA can be detected. One color assay, termed protein induced fluorescence enhancement enables detection of unlabeled protein binding and diffusion on a singly labeled RNA. Two-color Fluorescence Resonance Energy Transfer (FRET) in which labeled dsRBPs is applied to labeled RNA, allows for probing the motion of protein along the RNA axis. Three color FRET reports on the diffusion movement of dsRBPs from one to the other end of RNA. The single molecule pull down assay provides an opportunity to collect dsRBPs from mammalian cells and examine the protein-RNA interaction at single molecule platform.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , RNA, Double-Stranded/chemistry , RNA-Binding Proteins/chemistry , Single Molecule Imaging/methods , Animals , Diffusion , Mammals , RNA Interference , RNA, Double-Stranded/genetics , RNA-Binding Proteins/genetics
10.
Biochem Biophys Res Commun ; 480(2): 187-193, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27743889

ABSTRACT

LGP2 and MDA5 cooperate to detect viral RNA in the cytoplasm of Picornavirus-infected cells and activate innate immune responses. To further define regulatory components of RNA recognition by LGP2/MDA5, a yeast two-hybrid screen was used to identify LGP2-interacting proteins. The screening has identified the TAR-RNA binding protein (TRBP), which is known to be an essential factor for RNA interference (RNAi). Immuno-precipitation experiments demonstrated that TRBP interacted specifically with LGP2 but not with related RIG-I-like receptors, RIG-I or MDA5. siRNA knockdown experiments indicate that TRBP is important for Cardiovirus-triggered interferon responses, but TRBP is not involved in Sendai virus-triggered interferon response that is mediated mainly by RIG-I. To support functional interaction with LGP2, overexpressed TRBP increased Cardiovirus-triggered interferon promoter activity only when LGP2 and MDA5 are co-expressed but not MDA5 alone. Together, our findings illustrate a possible connection between an RNAi-regulatory factor and antiviral RNA recognition that is specifically required for a branch of the virus induced innate immune response.


Subject(s)
Cardiovirus Infections/metabolism , Host-Pathogen Interactions , RNA-Binding Proteins/metabolism , Animals , Cardiovirus/pathogenicity , Cardiovirus Infections/immunology , Chlorocebus aethiops , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , HEK293 Cells , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferon-beta/genetics , Mice , Promoter Regions, Genetic , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Small Interfering , RNA-Binding Proteins/genetics , Receptors, Immunologic , Sendai virus/pathogenicity , Vero Cells
11.
RNA Biol ; 12(2): 123-35, 2015.
Article in English | MEDLINE | ID: mdl-25668122

ABSTRACT

Several proteins and RNAs expressed by mammalian viruses have been reported to interfere with RNA interference (RNAi) activity. We investigated the ability of the HIV-1-encoded RNA elements Trans-Activation Response (TAR) and Rev-Response Element (RRE) to alter RNAi. MicroRNA let7-based assays showed that RRE is a potent suppressor of RNAi activity, while TAR displayed moderate RNAi suppression. We demonstrate that RRE binds to TAR-RNA Binding Protein (TRBP), an essential component of the RNA Induced Silencing Complex (RISC). The binding of TAR and RRE to TRBP displaces small interfering (si)RNAs from binding to TRBP. Several stem-deleted RRE mutants lost their ability to suppress RNAi activity, which correlated with a reduced ability to compete with siRNA-TRBP binding. A lentiviral vector expressing TAR and RRE restricted RNAi, but RNAi was restored when Rev or GagPol were coexpressed. Adenoviruses are restricted by RNAi and encode their own suppressors of RNAi, the Virus-Associated (VA) RNA elements. RRE enhanced the replication of wild-type and VA-deficient adenovirus. Our work describes RRE as a novel suppressor of RNAi that acts by competing with siRNAs rather than by disrupting the RISC. This function is masked in lentiviral vectors co-expressed with viral proteins and thus will not affect their use in gene therapy. The potent RNAi suppressive effects of RRE identified in this study could be used to enhance the expression of RNAi restricted viruses used in oncolysis such as adenoviruses.


Subject(s)
Genes, env , HIV Long Terminal Repeat , HIV-1/genetics , RNA Interference , RNA-Binding Proteins/genetics , Adenoviridae/genetics , Adenoviridae/metabolism , Binding, Competitive , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , HIV-1/metabolism , HeLa Cells , Host-Pathogen Interactions , Humans , Jurkat Cells , Lentivirus/genetics , Lentivirus/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nucleic Acid Conformation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , pol Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/metabolism
12.
Nanomedicine ; 11(6): 1455-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25720988

ABSTRACT

Efficiency of systemically delivered siRNA in gene silencing is compromised due to lack of target-specific delivery and rapid clearance of siRNA by in vivo elimination pathways. We designed a fusion protein consisting of a dsRNA binding domain of transactivation response RNA binding protein (TRBP2) fused to ErbB2 binding affibody (AF) for target specific delivery of siRNA. Designated as TRAF, the fusion protein is stable and binds efficiently and specifically to siRNA, forming homogenous non-aggregated and nuclease-resistant particles that efficiently and selectively transport siRNA into HER-2 overexpressing cancer cells and tissues. Administration of siRNA by TRAF into cells resulted in significant silencing of chosen genes involved in cell proliferation viz. AURKB and ErbB2. Noticeably, intravenous administration of TRAF:siRNA against these genes resulted in remarkable tumor suppression in the SK-OV-3 xenograft mouse model. Our results establish the potential of engineered proteins for specific and systemic delivery of siRNA for cancer therapy. FROM THE CLINICAL EDITOR: The use of siRNA in one of many novel treatments in cancer therapy. However, a major challenge for using siRNA is the lack of specificity and rapid RNA clearance. In this article, the authors designed a tumor targeting fusion protein, which can deliver siRNA specifically. In the experimental xenograft model, it was shown that intravenous administration of this resulted in significant tumor suppression. The results seem to hold promise in future clinical studies.


Subject(s)
RNA, Small Interfering/administration & dosage , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/genetics , Animals , Aurora Kinase B/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Silencing , Genes, erbB-2 , Humans , Mice , Mice, Inbred BALB C , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
13.
Biochim Biophys Acta ; 1833(12): 2673-2681, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23827255

ABSTRACT

RNAi is a highly conserved mechanism in almost every eukaryote with a few exceptions including the model organism Saccharomyces cerevisiae. A recent study showed that the introduction of the two core components of canonical RNAi systems, Argonaute and Dicer, from another budding yeast, Saccharomyces castellii, restores RNAi in S. cerevisiae. We report here that a functional RNAi system can be reconstituted in yeast with the introduction of only S. castellii Dicer and human Argonaute2. Interestingly, whether or not TRBP2 was present, human Dicer was unable to restore RNAi with either S. castellii or human Argonaute. Contrary to previous reports, we find that human Dicer, TRBP2 and Argonaute2 are not sufficient to reconstitute RNAi in yeast when bona fide RNAi precursors are co-expressed. We and others have previously reported that Hsp90 regulates conformational changes in human and Drosophila Argonautes required to accommodate the loading of dsRNA duplexes. Here we show that the activities of both human and S. castellii Argonaute are subject to Hsp90 regulation in S. cerevisiae. In summary, our results suggest that regulation of the RNAi machinery by Hsp90 may have evolved at the same time as ancestral RNAi.


Subject(s)
Evolution, Molecular , HSP90 Heat-Shock Proteins/metabolism , RNA Interference , Saccharomyces cerevisiae/metabolism , Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , Fluorescence , Gene Silencing , Green Fluorescent Proteins/metabolism , Humans , Macrolides/pharmacology , Protein Structure, Tertiary , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/metabolism
14.
Toxicol Sci ; 191(2): 332-342, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36453846

ABSTRACT

Benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE) leads to dysfunctions of human trophoblast cells and further induces miscarriage. In our previous study, we have found that lnc-HZ03 and miR-hz03 are upregulated in BPDE-exposed human trophoblast cells and in recurrent miscarriage tissues; and the upregulated miR-hz03 caused by lnc-HZ03 further promotes the apoptosis of human trophoblast cells and induces miscarriage. However, how lnc-HZ03 upregulates miR-hz03 is completely unknown. In this study, we find that lnc-HZ03 upregulates the expression level of a transcription factor TFIID (a TATA-binding protein) and promotes TFIID-mediated transactivation response element RNA-binding protein (TRBP) transcription. Subsequently, the upregulated TRBP promotes the maturation of miR-hz03 by splicing its precursor pre-miR-hz03 in human trophoblast cells. In BPDE-exposed trophoblast cells or in recurrent miscarriage tissues, lnc-HZ03 was upregulated, which accelerates the TFIID-mediated TRBP transcription and thus promotes TRBP-mediated miR-HZ03 maturation. Subsequently, the upregulated miR-hz03 further promotes the apoptosis of human trophoblast cells and induces miscarriage. This work provides new insights into the regulation of miRNA expression levels by lncRNAs in BPDE-exposed human trophoblast cells.


Subject(s)
Abortion, Habitual , MicroRNAs , RNA, Long Noncoding , Pregnancy , Female , Humans , Trophoblasts/metabolism , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide , MicroRNAs/genetics , MicroRNAs/metabolism , Abortion, Habitual/metabolism , Transcription Factor TFIID/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
15.
Cell Stress Chaperones ; 27(1): 11-25, 2022 01.
Article in English | MEDLINE | ID: mdl-34719748

ABSTRACT

Cells respond to stress through adaptive mechanisms that limit cellular damage and prevent cell death. MicroRNAs act as regulators of stress responses and stress can impact the functioning of miRNA biogenesis pathways. We were interested in the effect that severe proteotoxic stress capable of inducing apoptosis may have on miRNA biogenesis and the impact of the molecular chaperone protein HSP70 under these conditions. We found that the miRNA processing enzymes Drosha and Dicer and their accessory proteins DGCR8 and TRBP2 are cleaved by caspases in apoptotic cells. Overexpression of HSP70 prevented caspase activation and the degradation of these processing proteins. Caspase cleavage of TRBP2 was mapped to amino acid 234 which separates the two dsRNA-binding domains from the C-terminal Dicer interacting domain. Overexpression of TRBP2 was found to increase miRNA maturation, while expression of either of the fragments generated by caspase cleavage impaired maturation. These results indicate that inactivation of miRNA biogenesis is a critical feature of apoptosis and that cleavage of TRBP2, rather than simply a loss of function, serves to create positive acting inhibitors of pre-miRNA maturation.


Subject(s)
MicroRNAs , RNA-Binding Proteins , Caspases/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hot Temperature , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
16.
Front Microbiol ; 13: 859420, 2022.
Article in English | MEDLINE | ID: mdl-35558132

ABSTRACT

Influenza A virus (IAV), one of the most prevalent respiratory diseases, causes pandemics around the world. The multifunctional non-structural protein 1 (NS1) of IAV is a viral antagonist that suppresses host antiviral response. However, the mechanism by which NS1 modulates the RNA interference (RNAi) pathway remains unclear. Here, we identified interactions between NS1 proteins of Influenza A/PR8/34 (H1N1; IAV-PR8) and Influenza A/WSN/1/33 (H1N1; IAV-WSN) and Dicer's cofactor TAR-RNA binding protein (TRBP). We found that the N-terminal RNA binding domain (RBD) of NS1 and the first two domains of TRBP protein mediated this interaction. Furthermore, two amino acid residues (Arg at position 38 and Lys at position 41) in NS1 were essential for the interaction. We generated TRBP knockout cells and found that NS1 instead of NS1 mutants (two-point mutations within NS1, R38A/K41A) inhibited the process of microRNA (miRNA) maturation by binding with TRBP. PR8-infected cells showed masking of short hairpin RNA (shRNA)-mediated RNAi, which was not observed after mutant virus-containing NS1 mutation (R38A/K41A, termed PR8/3841) infection. Moreover, abundant viral small interfering RNAs (vsiRNAs) were detected in vitro and in vivo upon PR8/3841 infection. We identify, for the first time, the interaction between NS1 and TRBP that affects host RNAi machinery.

17.
Int J Biochem Cell Biol ; 137: 106030, 2021 08.
Article in English | MEDLINE | ID: mdl-34174402

ABSTRACT

Transactivation response element RNA-binding protein (TRBP or TARBP2) originally identified as a pro-viral cellular protein in human immunodeficiency virus (HIV) replication is also a regulator of microRNA biogenesis and cellular stress response. TRBP inhibits the catalytic activity of interferon-induced double-stranded RNA (dsRNA)-activated protein kinase (PKR) during viral infections and cell stress thereby regulating stress-induced signaling pathways. During cellular stress, PKR is catalytically activated transiently by its protein activator PACT and TRBP inhibits PKR to bring about a timely cellular recovery. We have previously established that TRBP phosphorylated after oxidative stress binds to and inhibits PKR more efficiently promoting cell survival. In this study, we investigated if phosphorylation of TRBP enhances its interaction with PACT to bring about additional PKR inhibition. Our data establishes that phosphorylation of TRBP has no effect on PACT-TRBP interaction and TRBP's inhibitory actions on PKR are mediated exclusively by its enhanced interaction with PKR. Cells lacking TRBP are more sensitive to apoptosis in response to oxidative stress and show persistent PKR activation. These results establish that PKR inhibition by stress-induced TRBP phosphorylation occurs by its direct binding to PKR and is important for preventing apoptosis due to sustained PKR activation.


Subject(s)
Apoptosis , Oxidative Stress , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , eIF-2 Kinase/metabolism , Animals , HeLa Cells , Humans , Mice , Phosphorylation , RNA, Double-Stranded/genetics , RNA-Binding Proteins/genetics , Signal Transduction , eIF-2 Kinase/genetics
18.
Neurochem Int ; 151: 105210, 2021 12.
Article in English | MEDLINE | ID: mdl-34695450

ABSTRACT

A growing number of evidence suggests that altered microRNA network in the brain contributes to the risk of Alzheimer's disease(AD). Dicer1 is a type III riboendonuclease which cleaves pre-microRNA into functional microRNA. Reduction of Dicer1 or Dicer1 mutation has been involved in cancer, aging or age-related macular degeneration. Recently, we found a possible link between Dicer1 and AD. In particular, Dicer1 protein and Dicer1 mRNA is reduced in the hippocampus and the cortex of an animal model of AD and exposure to Aß42 oligomer(AßO) longer than 6 h reduces the transcription of Dicer1 gene in neuron, via depletion of NF-E2-related factor-2. In this study, exposure to AßO at shorter time increased Dicer1 protein in neuron in a dose-dependent mode; but the mRNA level remained unaltered. Under this treatment regime,AßO reduced phosphorylation level of Dicer1 and of its binding partner, transactivation response element RNA-binding protein(TRBP). Addition of a JNK inhibitor,SP600125, or an ERK inhibitor,U0126, further increased Dicer1 protein compared to Aßo treatment alone, with simultaneaous reduction of phospho-Dicer1, but with different effects on phospho-TRBP. Finally, an inhibitor of calcineurin,FK506, further increased Dicer1 protein compared to Aßo treatment alone. Thus, phosphorylation of Dicer1 and TRBP was determined by mitogen activated protein kinases JNK,ERK, and protein phosphatase 2B(calcineurin) which together determined Dicer1 stability. In summary, reduced phosphorylation of Dicer1 accounted for the rapid induction of Dicer1 by AßO. This study highlights a novel way by which AßO regulates Dicer1.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , DEAD-box RNA Helicases/metabolism , Neurons/metabolism , Ribonuclease III/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Brain/drug effects , DEAD-box RNA Helicases/genetics , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Phosphorylation , Ribonuclease III/genetics
19.
Front Cardiovasc Med ; 8: 773314, 2021.
Article in English | MEDLINE | ID: mdl-34957257

ABSTRACT

Various stresses, including pressure overload and myocardial stretch, can trigger cardiac remodeling and result in heart diseases. The disorders are associated with high risk of morbidity and mortality and are among the major health problems in the world. MicroRNAs, a class of ~22nt-long small non-coding RNAs, have been found to participate in regulating heart development and function. One of them, miR-208a, a cardiac-specific microRNA, plays key role(s) in modulating gene expression in the heart, and is involved in a broad array of processes in cardiac pathogenesis. Genetic deletion or pharmacological inhibition of miR-208a in rodents attenuated stress-induced cardiac hypertrophy and remodeling. Transgenic expression of miR-208a in the heart was sufficient to cause hypertrophic growth of cardiomyocytes. miR-208a is also a key regulator of cardiac conduction system, either deletion or transgenic expression of miR-208a disturbed heart electrophysiology and could induce arrhythmias. In addition, miR-208a appeared to assist in regulating the expression of fast- and slow-twitch myofiber genes in the heart. Notably, this heart-specific miRNA could also modulate the "endocrine" function of cardiac muscle and govern the systemic energy homeostasis in the whole body. Despite of the critical roles, the underlying regulatory networks involving miR-208a are still elusive. Here, we summarize the progress made in understanding the function and mechanisms of this important miRNA in the heart, and propose several topics to be resolved as well as the hypothetical answers. We speculate that miR-208a may play diverse and even opposite roles by being involved in distinct molecular networks depending on the contexts. A deeper understanding of the precise mechanisms of its action under the conditions of cardiac homeostasis and diseases is needed. The clinical implications of miR-208a are also discussed.

20.
Virus Res ; 298: 198404, 2021 06.
Article in English | MEDLINE | ID: mdl-33775754

ABSTRACT

MicroRNAs are emerging as critical endogenous regulators of gene function. Aberrant regulation of microRNAs is associated with various human diseases, most importantly cancer. MicroRNA-122 (miR-122), a liver-specific microRNA, has been implicated in the control of hepatitis C virus (HCV) RNA replication and its response to interferon (IFN) in human hepatoma cells. Here, we report that daidzein, a naturally occurring plant isoflavone, inhibits HCV replication and enhances the antiviral effect of IFN-α on HCV therapy by decreasing microRNA-122 levels in vitro without significantly affecting cell growth. Moreover, daidzein was found to inhibit the expression of miR-122 and miR-21 by down-regulating the expression of TRBP, indicating that daidzein is possibly a general inhibitor of the miRNA pathway. Thus, daidzein provides new insights for drug discovery and HCV prevention.


Subject(s)
Hepatitis C , Isoflavones , MicroRNAs , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Hepacivirus/physiology , Hepatitis C/genetics , Humans , Interferon-alpha/metabolism , Isoflavones/metabolism , Isoflavones/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL