Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.947
Filter
1.
J Comput Aided Mol Des ; 38(1): 25, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39014124

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) are pivotal RNA-editing enzymes responsible for converting adenosine to inosine within double-stranded RNA (dsRNA). Dysregulation of ADAR1 editing activity, often arising from genetic mutations, has been linked to elevated interferon levels and the onset of autoinflammatory diseases. However, understanding the molecular underpinnings of this dysregulation is impeded by the lack of an experimentally determined structure for the ADAR1 deaminase domain. In this computational study, we utilized homology modeling and the AlphaFold2 to construct structural models of the ADAR1 deaminase domain in wild-type and two pathogenic variants, R892H and Y1112F, to decipher the structural impact on the reduced deaminase activity. Our findings illuminate the critical role of structural complementarity between the ADAR1 deaminase domain and dsRNA in enzyme-substrate recognition. That is, the relative position of E1008 and K1120 must be maintained so that they can insert into the minor and major grooves of the substrate dsRNA, respectively, facilitating the flipping-out of adenosine to be accommodated within a cavity surrounding E912. Both amino acid replacements studied, R892H at the orthosteric site and Y1112F at the allosteric site, alter K1120 position and ultimately hinder substrate RNA binding.


Subject(s)
Adenosine Deaminase , Molecular Dynamics Simulation , RNA-Binding Proteins , Adenosine Deaminase/chemistry , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Humans , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mutation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , Protein Conformation , RNA Editing
2.
Postepy Biochem ; 70(1): 57-61, 2024 05 23.
Article in English | MEDLINE | ID: mdl-39016229

ABSTRACT

MicroRNAs (miRNAs) are generated from stem-loop-structured double-stranded RNA precursors by the consecutive action of the two RNase III-type endoribonuclease Drosha and Dicer. However, such structures are very common on cellular transcripts and specific features have evolved that guide and regulate processing of stem-loop-structured hairpins into mature and functional miRNAs. These features include sequence motifs and local RNA structures but also trans-acting factors such as RNA binding proteins. The menu of features required for miRNA biogenesis is summarized in this review.


Subject(s)
MicroRNAs , Ribonuclease III , MicroRNAs/metabolism , MicroRNAs/genetics , Humans , Animals , Ribonuclease III/metabolism , Ribonuclease III/genetics , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , RNA Precursors/metabolism , RNA Precursors/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Nucleic Acid Conformation
3.
Proc Natl Acad Sci U S A ; 121(31): e2409233121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39047046

ABSTRACT

Invertebrates mainly rely on sequence-specific RNA interference (RNAi) to resist viral infections. Increasing studies show that double-stranded RNA (dsRNA) can induce sequence-independent protection and that Dicer-2, the key RNAi player that cleaves long dsRNA into small interfering RNA (siRNA), is necessary for this protection. However, how this protection occurs remains unknown. Herein, we report that it is caused by adenosine triphosphate (ATP)-hydrolysis accompanying the dsRNA-cleavage. Dicer-2 helicase domain is ATP-dependent; therefore, the cleavage consumes ATP. ATP depletion activates adenosine monophosphate-activated protein kinase (Ampk) and induces nuclear localization of Fork head box O (FoxO), a key transcriptional factor for dsRNA-induced genes. siRNAs that do not require processing cannot activate the transcriptional response. This study reveals a unique nonspecific antiviral mechanism other than the specific RNAi in shrimp. This mechanism is functionally similar to, but mechanistically different from, the dsRNA-activated antiviral response in vertebrates and suggests an interesting evolution of innate antiviral immunity.


Subject(s)
AMP-Activated Protein Kinases , Adenosine Triphosphate , RNA, Double-Stranded , Ribonuclease III , Animals , RNA, Double-Stranded/metabolism , Ribonuclease III/metabolism , Ribonuclease III/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Immunity, Innate , Transcription, Genetic
4.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38955468

ABSTRACT

In addition to mitochondrial DNA, mitochondrial double-stranded RNA (mtdsRNA) is exported from mitochondria. However, specific channels for RNA transport have not been demonstrated. Here, we begin to characterize channel candidates for mtdsRNA export from the mitochondrial matrix to the cytosol. Down-regulation of SUV3 resulted in the accumulation of mtdsRNAs in the matrix, whereas down-regulation of PNPase resulted in the export of mtdsRNAs to the cytosol. Targeting experiments show that PNPase functions in both the intermembrane space and matrix. Strand-specific sequencing of the double-stranded RNA confirms the mitochondrial origin. Inhibiting or down-regulating outer membrane proteins VDAC1/2 and BAK/BAX or inner membrane proteins PHB1/2 strongly attenuated the export of mtdsRNAs to the cytosol. The cytosolic mtdsRNAs subsequently localized to large granules containing the stress protein TIA-1 and activated the type 1 interferon stress response pathway. Abundant mtdsRNAs were detected in a subset of non-small-cell lung cancer cell lines that were glycolytic, indicating relevance in cancer biology. Thus, we propose that mtdsRNA is a new damage-associated molecular pattern that is exported from mitochondria in a regulated manner.


Subject(s)
Cytosol , Mitochondria , Prohibitins , RNA, Double-Stranded , RNA, Mitochondrial , Humans , Cytosol/metabolism , Mitochondria/metabolism , RNA, Double-Stranded/metabolism , RNA, Mitochondrial/metabolism , RNA, Mitochondrial/genetics , Cell Line, Tumor , Repressor Proteins/metabolism , Repressor Proteins/genetics , RNA Transport , Exoribonucleases/metabolism , Exoribonucleases/genetics , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channel 1/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Mitochondrial Proteins
5.
Proc Natl Acad Sci U S A ; 121(29): e2402126121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38980902

ABSTRACT

Upon sensing viral RNA, mammalian RIG-I-like receptors (RLRs) activate downstream signals using caspase activation and recruitment domains (CARDs), which ultimately promote transcriptional immune responses that have been well studied. In contrast, the downstream signaling mechanisms for invertebrate RLRs are much less clear. For example, the Caenorhabditis elegans RLR DRH-1 lacks annotated CARDs and up-regulates the distinct output of RNA interference. Here, we found that similar to mammal RLRs, DRH-1 signals through two tandem CARDs (2CARD) to induce a transcriptional immune response. Expression of DRH-1(2CARD) alone in the intestine was sufficient to induce immune gene expression, increase viral resistance, and promote thermotolerance, a phenotype previously associated with immune activation in C. elegans. We also found that DRH-1 is required in the intestine to induce immune gene expression, and we demonstrate subcellular colocalization of DRH-1 puncta with double-stranded RNA inside the cytoplasm of intestinal cells upon viral infection. Altogether, our results reveal mechanistic and spatial insights into antiviral signaling in C. elegans, highlighting unexpected parallels in RLR signaling between C. elegans and mammals.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Signal Transduction , Animals , Caenorhabditis elegans/immunology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Signal Transduction/immunology , Intestines/immunology , Intestines/virology , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/immunology , Immunity, Innate , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , RNA, Viral/immunology , RNA, Viral/metabolism , RNA, Viral/genetics
6.
PLoS Pathog ; 20(7): e1012320, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39012849

ABSTRACT

Apoptosis, a major form of programmed cell death, is an essential component of host defense against invading intracellular pathogens. Viruses encode inhibitors of apoptosis to evade host responses during infection, and to support their own replication and survival. Therefore, hosts and their viruses are entangled in a constant evolutionary arms race to control apoptosis. Until now, apoptosis in the context of the antiviral immune system has been almost exclusively studied in vertebrates. This limited phyletic sampling makes it impossible to determine whether a similar mechanism existed in the last common ancestor of animals. Here, we established assays to probe apoptosis in the sea anemone Nematostella vectensis, a model species of Cnidaria, a phylum that diverged approximately 600 million years ago from the rest of animals. We show that polyinosinic:polycytidylic acid (poly I:C), a synthetic long double-stranded RNA mimicking viral RNA and a primary ligand for the vertebrate RLR melanoma differentiation-associated protein 5 (MDA5), is sufficient to induce apoptosis in N. vectensis. Furthermore, at the transcriptomic level, apoptosis related genes are significantly enriched upon poly(I:C) exposure in N. vectensis as well as bilaterian invertebrates. Our phylogenetic analysis of caspase family genes in N. vectensis reveals conservation of all four caspase genes involved in apoptosis in mammals and revealed a cnidarian-specific caspase gene which was strongly upregulated. Altogether, our findings suggest that apoptosis in response to a viral challenge is a functionally conserved mechanism that can be traced back to the last common ancestor of Bilateria and Cnidaria.


Subject(s)
Apoptosis , RNA, Double-Stranded , Sea Anemones , Animals , Sea Anemones/genetics , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , Phylogeny , Poly I-C/pharmacology , Cnidaria/genetics , Biological Evolution
7.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062980

ABSTRACT

Mitochondrial stress, resulting from dysfunction and proteostasis disturbances, triggers the mitochondrial unfolded protein response (UPRMT), which activates gene encoding chaperones and proteases to restore mitochondrial function. Although ATFS-1 mediates mitochondrial stress UPRMT induction in C. elegans, the mechanisms relaying mitochondrial stress signals to the nucleus in mammals remain poorly defined. Here, we explored the role of protein kinase R (PKR), an eIF2α kinase activated by double-stranded RNAs (dsRNAs), in mitochondrial stress signaling. We found that UPRMT does not occur in cells lacking PKR, indicating its crucial role in this process. Mechanistically, we observed that dsRNAs accumulate within mitochondria under stress conditions, along with unprocessed mitochondrial transcripts. Furthermore, we demonstrated that accumulated mitochondrial dsRNAs in mouse embryonic fibroblasts (MEFs) deficient in the Bax/Bak channels are not released into the cytosol and do not induce the UPRMT upon mitochondrial stress, suggesting a potential role of the Bax/Bak channels in mediating the mitochondrial stress response. These discoveries enhance our understanding of how cells maintain mitochondrial integrity, respond to mitochondrial dysfunction, and communicate stress signals to the nucleus through retrograde signaling. This knowledge provides valuable insights into prospective therapeutic targets for diseases associated with mitochondrial stress.


Subject(s)
Mitochondria , RNA, Double-Stranded , Unfolded Protein Response , eIF-2 Kinase , Animals , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Mitochondria/metabolism , RNA, Double-Stranded/metabolism , Mice , Stress, Physiological , Signal Transduction , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics , Fibroblasts/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , Humans
8.
PLoS One ; 19(7): e0307976, 2024.
Article in English | MEDLINE | ID: mdl-39074084

ABSTRACT

White spot syndrome virus (WSSV) presents a considerable peril to the aquaculture sector, leading to notable financial consequences on a global scale. Previous studies have identified hub proteins, including WSSV051 and WSSV517, as essential binding elements in the protein interaction network of WSSV. This work further investigates the functional structures and potential applications of WSSV hub complexes in managing WSSV infection. Using computational methodologies, we have successfully generated comprehensive three-dimensional (3D) representations of hub proteins along with their three mutual binding counterparts, elucidating crucial interaction locations. The results of our study indicate that the WSSV051 hub protein demonstrates higher binding energy than WSSV517. Moreover, a unique motif, denoted as "S-S-x(5)-S-x(2)-P," was discovered among the binding proteins. This pattern perhaps contributes to the detection of partners by the hub proteins of WSSV. An antiviral strategy targeting WSSV hub proteins was demonstrated through the oral administration of dual hub double-stranded RNAs to the black tiger shrimp, Penaeus monodon, followed by a challenge assay. The findings demonstrate a decrease in shrimp mortality and a cessation of WSSV multiplication. In conclusion, our research unveils the structural features and dynamic interactions of hub complexes, shedding light on their significance in the WSSV protein network. This highlights the potential of hub protein-based interventions to mitigate the impact of WSSV infection in aquaculture.


Subject(s)
Penaeidae , Viral Proteins , White spot syndrome virus 1 , Animals , White spot syndrome virus 1/physiology , Penaeidae/virology , Viral Proteins/metabolism , Viral Proteins/chemistry , Models, Molecular , Protein Binding , Amino Acid Sequence , RNA, Double-Stranded/metabolism , Protein Interaction Maps , Aquaculture
9.
Proc Natl Acad Sci U S A ; 121(25): e2322765121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38865263

ABSTRACT

Antiviral RNA interference (RNAi) is conserved from yeasts to mammals. Dicer recognizes and cleaves virus-derived double-stranded RNA (dsRNA) and/or structured single-stranded RNA (ssRNA) into small-interfering RNAs, which guide effector Argonaute to homologous viral RNAs for digestion and inhibit virus replication. Thus, Argonaute is believed to be essential for antiviral RNAi. Here, we show Argonaute-independent, Dicer-dependent antiviral defense against dsRNA viruses using Cryphonectria parasitica (chestnut blight fungus), which is a model filamentous ascomycetous fungus and hosts a variety of viruses. The fungus has two dicer-like genes (dcl1 and dcl2) and four argonaute-like genes (agl1 to agl4). We prepared a suite of single to quadruple agl knockout mutants with or without dcl disruption. We tested these mutants for antiviral activities against diverse dsRNA viruses and ssRNA viruses. Although both DCL2 and AGL2 worked as antiviral players against some RNA viruses, DCL2 without argonaute was sufficient to block the replication of other RNA viruses. Overall, these results indicate the existence of a Dicer-alone defense and different degrees of susceptibility to it among RNA viruses. We discuss what determines the great difference in susceptibility to the Dicer-only defense.


Subject(s)
RNA Viruses , Ribonuclease III , Ribonuclease III/metabolism , Ribonuclease III/genetics , RNA Viruses/immunology , RNA Viruses/genetics , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Ascomycota/virology , RNA Interference , Virus Replication/genetics , RNA, Viral/metabolism , RNA, Viral/genetics , Fungal Proteins/metabolism , Fungal Proteins/genetics , RNA, Double-Stranded/metabolism
10.
Biochem Soc Trans ; 52(3): 1131-1148, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38884803

ABSTRACT

The RIG-I-like receptors (RLRs), comprising retinoic acid-inducible gene I (RIG-I), melanoma differentiation-associated gene 5 (MDA5), and laboratory of genetics and physiology 2 (LGP2), are pattern recognition receptors belonging to the DExD/H-box RNA helicase family of proteins. RLRs detect viral RNAs in the cytoplasm and respond by initiating a robust antiviral response that up-regulates interferon and cytokine production. RIG-I and MDA5 complement each other by recognizing different RNA features, and LGP2 regulates their activation. RIG-I's multilayered RNA recognition and proofreading mechanisms ensure accurate viral RNA detection while averting harmful responses to host RNAs. RIG-I's C-terminal domain targets 5'-triphosphate double-stranded RNA (dsRNA) blunt ends, while an intrinsic gating mechanism prevents the helicase domains from non-specifically engaging with host RNAs. The ATPase and RNA translocation activity of RIG-I adds another layer of selectivity by minimizing the lifetime of RIG-I on non-specific RNAs, preventing off-target activation. The versatility of RIG-I's ATPase function also amplifies downstream signaling by enhancing the signaling domain (CARDs) exposure on 5'-triphosphate dsRNA and promoting oligomerization. In this review, we offer an in-depth understanding of the mechanisms RIG-I uses to facilitate viral RNA sensing and regulate downstream activation of the immune system.


Subject(s)
DEAD Box Protein 58 , Immunity, Innate , RNA, Viral , Receptors, Immunologic , Humans , RNA, Viral/metabolism , DEAD Box Protein 58/metabolism , Receptors, Immunologic/metabolism , Animals , RNA, Double-Stranded/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , DEAD-box RNA Helicases/metabolism , RNA Helicases/metabolism
11.
Parasit Vectors ; 17(1): 255, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38863029

ABSTRACT

BACKGROUND: RNA interference (RNAi) is a target-specific gene silencing method that can be used to determine gene functions and investigate host-pathogen interactions, as well as facilitating the development of ecofriendly pesticides. Commercially available transfection reagents (TRs) can improve the efficacy of RNAi. However, we currently lack a product and protocol for the transfection of insect cell lines with long double-stranded RNA (dsRNA). METHODS: We used agarose gel electrophoresis to determine the capacity of eight TRs to form complexes with long dsRNA. A CellTiter-Glo assay was then used to assess the cytotoxicity of the resulting lipoplexes. We also measured the cellular uptake of dsRNA by fluorescence microscopy using the fluorophore Cy3 as a label. Finally, we analyzed the TRs based on their transfection efficacy and compared the RNAi responses of Aedes albopictus C6/36 and U4.4 cells by knocking down an mCherry reporter Semliki Forest virus in both cell lines. RESULTS: The TRs from Biontex (K4, Metafectene Pro, and Metafectene SI+) showed the best complexing capacity and the lowest dsRNA:TR ratio needed for complete complex formation. Only HiPerFect was unable to complex the dsRNA completely, even at a ratio of 1:9. Most of the complexes containing mCherry-dsRNA were nontoxic at 2 ng/µL, but Lipofectamine 2000 was toxic at 1 ng/µL in U4.4 cells and at 2 ng/µL in C6/36 cells. The transfection of U4.4 cells with mCherry-dsRNA/TR complexes achieved significant knockdown of the virus reporter. Comparison of the RNAi response in C6/36 and U4.4 cells suggested that C6/36 cells lack the antiviral RNAi response because there was no significant knockdown of the virus reporter in any of the treatments. CONCLUSIONS: C6/36 cells have an impaired RNAi response as previously reported. This investigation provides valuable information for future RNAi experiments by showing how to mitigate the adverse effects attributed to TRs. This will facilitate the judicious selection of TRs and transfection conditions conducive to RNAi research in mosquitoes.


Subject(s)
Aedes , RNA Interference , RNA, Double-Stranded , Transfection , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Animals , Cell Line , Aedes/genetics , Gene Silencing , Semliki forest virus/genetics , Semliki forest virus/drug effects
12.
Bull Exp Biol Med ; 176(6): 751-755, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38896322

ABSTRACT

The removal of double-stranded RNA (dsRNA) contaminants during in vitro mRNA synthesis is one of the technological problems to be solved. Apparently, these contaminants are the result of the T7 RNA polymerase side activity. In this study, we used a modified method of mRNA purification based on the selective binding of dsRNA to cellulose in ethanol-containing buffer. It was shown both in vivo and in vitro that the cellulose-purified mRNA preparation leads neither to activation of the lymphocyte inflammatory marker CD69 nor to increased release of IFNα in mice, and does not contain impurities detectable by antibodies to dsRNA.


Subject(s)
RNA, Double-Stranded , RNA, Messenger , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Mice , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, CD/genetics , Antigens, CD/metabolism , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-alpha/biosynthesis , Viral Proteins/metabolism , Viral Proteins/genetics
13.
Cell Rep ; 43(6): 114287, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38823018

ABSTRACT

Viral infection triggers several double-stranded RNA (dsRNA) sensors that lead to changes in gene expression in the cell. One of these sensors activates an endonuclease, ribonuclease L (RNase L), that cleaves single-stranded RNA. However, how the resultant widespread RNA fragmentation affects gene expression is not fully understood. Here, we show that this fragmentation induces the ribotoxic stress response via ZAKα, potentially through stalled ribosomes and/or ribosome collisions. The p38 and JNK pathways that are activated as part of this response promote outcomes that inhibit the virus, such as programmed cell death. We also show that RNase L limits the translation of stress-responsive genes. Intriguingly, we found that the activity of the generic endonuclease, RNase A, recapitulates many of the same molecular phenotypes as activated RNase L, demonstrating how widespread RNA cleavage can evoke an antiviral program.


Subject(s)
Endoribonucleases , Immunity, Innate , Endoribonucleases/metabolism , Endoribonucleases/genetics , Humans , RNA Cleavage , Animals , RNA, Double-Stranded/metabolism , Mice , Ribonuclease, Pancreatic/metabolism
14.
Proc Natl Acad Sci U S A ; 121(26): e2322927121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38885386

ABSTRACT

RNA interference (RNAi) is more efficient in coleopteran insects than other insects. StaufenC (StauC), a coleopteran-specific double-stranded RNA (dsRNA)-binding protein, is required for efficient RNAi in coleopterans. We investigated the function of StauC in the intracellular transport of dsRNA into the cytosol, where dsRNA is digested by Dicer enzymes and recruited by Argonauts to RNA-induced silencing complexes. Confocal microscopy and cellular organelle fractionation studies have shown that dsRNA is trafficked through the endoplasmic reticulum (ER) in coleopteran Colorado potato beetle (CPB) cells. StauC is localized to the ER in CPB cells, and StauC-knockdown caused the accumulation of dsRNA in the ER and a decrease in the cytosol, suggesting that StauC plays a key role in the intracellular transport of dsRNA through the ER. Using immunoprecipitation, we showed that StauC is required for dsRNA interaction with ER proteins in the ER-associated protein degradation (ERAD) pathway, and these interactions are required for RNAi in CPB cells. These results suggest that StauC works with the ERAD pathway to transport dsRNA through the ER to the cytosol. This information could be used to develop dsRNA delivery methods aimed at improving RNAi.


Subject(s)
Coleoptera , Cytosol , Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum , RNA, Double-Stranded , RNA-Binding Proteins , Animals , Endoplasmic Reticulum/metabolism , RNA, Double-Stranded/metabolism , Cytosol/metabolism , Coleoptera/metabolism , Coleoptera/genetics , Endoplasmic Reticulum-Associated Degradation/physiology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Insect Proteins/metabolism , Insect Proteins/genetics , RNA Interference , Biological Transport
15.
Exp Mol Med ; 56(6): 1250-1262, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38871814

ABSTRACT

Alu elements are highly abundant primate-specific short interspersed nuclear elements that account for ~10% of the human genome. Due to their preferential location in gene-rich regions, especially in introns and 3' UTRs, Alu elements can exert regulatory effects on the expression of both host and neighboring genes. When two Alu elements with inverse orientations are positioned in close proximity, their transcription results in the generation of distinct double-stranded RNAs (dsRNAs), known as inverted Alu repeats (IRAlus). IRAlus are key immunogenic self-dsRNAs and post-transcriptional cis-regulatory elements that play a role in circular RNA biogenesis, as well as RNA transport and stability. Recently, IRAlus dsRNAs have emerged as regulators of transcription and activators of Z-DNA-binding proteins. The formation and activity of IRAlus can be modulated through RNA editing and interactions with RNA-binding proteins, and misregulation of IRAlus has been implicated in several immune-associated disorders. In this review, we summarize the emerging functions of IRAlus dsRNAs, the regulatory mechanisms governing IRAlus activity, and their relevance in the pathogenesis of human diseases.


Subject(s)
Alu Elements , Transcriptome , Humans , Alu Elements/genetics , Gene Expression Regulation , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Animals , RNA, Circular/genetics , Inverted Repeat Sequences , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA Editing
16.
Methods Mol Biol ; 2813: 235-244, 2024.
Article in English | MEDLINE | ID: mdl-38888782

ABSTRACT

Double-stranded RNA is produced by viruses during their replicative cycle. It is a potent immune modulator and indicator of viral infection within the body. Extracellular vesicles (EVs) are lipid-bound particles released from cells homeostatically. Recent studies have shown that a commercially available dsRNA, poly inosinic: poly cytidylic acid (poly IC), can be detected within EVs. This finding opens the door for studying EVs as (1) carriers for dsRNA and (2) indicators of viral infection. To study dsRNA-containing EVs, we must have reliable methods for producing, isolating, and detecting them. This chapter uses U937, a pro-monocytic, human myeloid leukemia cell line, as the EV producer following poly IC treatment, and an immunoblot using an anti-dsRNA antibody (J2) for detection. Two methods for isolating the EVs and two methods for isolating the RNA from these EVs are described. Together, these methods effectively produce, isolate, and detect long dsRNA from EVs.


Subject(s)
Extracellular Vesicles , Poly I-C , Humans , Extracellular Vesicles/metabolism , Poly I-C/pharmacology , U937 Cells , RNA, Double-Stranded/metabolism
17.
Cell Rep ; 43(7): 114400, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38935501

ABSTRACT

ADAR1-mediated RNA editing establishes immune tolerance to endogenous double-stranded RNA (dsRNA) by preventing its sensing, primarily by MDA5. Although deleting Ifih1 (encoding MDA5) rescues embryonic lethality in ADAR1-deficient mice, they still experience early postnatal death, and removing other MDA5 signaling proteins does not yield the same rescue. Here, we show that ablation of MDA5 in a liver-specific Adar knockout (KO) murine model fails to rescue hepatic abnormalities caused by ADAR1 loss. Ifih1;Adar double KO (dKO) hepatocytes accumulate endogenous dsRNAs, leading to aberrant transition to a highly inflammatory state and recruitment of macrophages into dKO livers. Mechanistically, progranulin (PGRN) appears to mediate ADAR1 deficiency-induced liver pathology, promoting interferon signaling and attracting epidermal growth factor receptor (EGFR)+ macrophages into dKO liver, exacerbating hepatic inflammation. Notably, the PGRN-EGFR crosstalk communication and consequent immune responses are significantly repressed in ADAR1high tumors, revealing that pre-neoplastic or neoplastic cells can exploit ADAR1-dependent immune tolerance to facilitate immune evasion.


Subject(s)
Adenosine Deaminase , ErbB Receptors , Hepatocytes , Interferon-Induced Helicase, IFIH1 , Liver , Macrophages , Mice, Knockout , Progranulins , Animals , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , ErbB Receptors/metabolism , Macrophages/metabolism , Macrophages/immunology , Progranulins/metabolism , Progranulins/genetics , Liver/metabolism , Liver/immunology , Liver/pathology , Hepatocytes/metabolism , Mice , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Signal Transduction , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice, Inbred C57BL , RNA, Double-Stranded/metabolism , RNA Editing
18.
Nature ; 631(8020): 432-438, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38898279

ABSTRACT

When mRNAs have been transcribed and processed in the nucleus, they are exported to the cytoplasm for translation. This export is mediated by the export receptor heterodimer Mex67-Mtr2 in the yeast Saccharomyces cerevisiae (TAP-p15 in humans)1,2. Interestingly, many long non-coding RNAs (lncRNAs) also leave the nucleus but it is currently unclear why they move to the cytoplasm3. Here we show that antisense RNAs (asRNAs) accelerate mRNA export by annealing with their sense counterparts through the helicase Dbp2. These double-stranded RNAs (dsRNAs) dominate export compared with single-stranded RNAs (ssRNAs) because they have a higher capacity and affinity for the export receptor Mex67. In this way, asRNAs boost gene expression, which is beneficial for cells. This is particularly important when the expression program changes. Consequently, the degradation of dsRNA, or the prevention of its formation, is toxic for cells. This mechanism illuminates the general cellular occurrence of asRNAs and explains their nuclear export.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus , Gene Expression Regulation, Fungal , RNA Transport , RNA, Antisense , RNA, Double-Stranded , RNA, Messenger , Saccharomyces cerevisiae , Cell Nucleus/metabolism , Cytoplasm/metabolism , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , RNA, Antisense/metabolism , RNA, Antisense/genetics , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
19.
J Agric Food Chem ; 72(22): 12508-12515, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38788129

ABSTRACT

Nanotechnology-based RNA interference (RNAi) offers a promising approach to pest control. However, current methods for producing RNAi nanopesticides are mainly implemented in a batch-to-batch manner, lacking consistent quality control. Herein, we present a microfluidic-based nanoplatform for RNA nanopesticide preparation using lipid nanoparticles (LNPs) as nanocarriers, taking advantage of the enhanced mass transfer and continuous processing capabilities of microfluidic technology. The dsRNA@LNPs were rapidly formed within seconds, which showed uniform size distribution, improved leaf wettability, and excellent dispersion properties. The delivery efficiency of dsRNA@LNPs was evaluated by targeting the chitin synthetase B (CHSB) gene ofSpodoptera exigua. The dsRNA@LNPs can effectively resist nuclease-rich midgut fluid degradation. Importantly, dsCHSB@LNPs exhibited increased mortality rates, significant reduction of larvae growth, and enhanced gene suppression efficiency. Therefore, a continuous nanoplatform for RNAi nanopesticide preparation is demonstrated by utilizing microfluidic technology, representing a new route to produce RNAi nanopesticides with enhanced quality control and might accelerate their practical applications.


Subject(s)
Larva , RNA Interference , RNA, Double-Stranded , Spodoptera , Animals , Spodoptera/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , Larva/growth & development , Larva/genetics , Nanoparticles/chemistry , Microfluidics/instrumentation , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Proteins/chemistry , Insect Control/methods
20.
Nat Commun ; 15(1): 4644, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821943

ABSTRACT

The SARS-CoV-2 viral infection transforms host cells and produces special organelles in many ways, and we focus on the replication organelles, the sites of replication of viral genomic RNA (vgRNA). To date, the precise cellular localization of key RNA molecules and replication intermediates has been elusive in electron microscopy studies. We use super-resolution fluorescence microscopy and specific labeling to reveal the nanoscopic organization of replication organelles that contain numerous vgRNA molecules along with the replication enzymes and clusters of viral double-stranded RNA (dsRNA). We show that the replication organelles are organized differently at early and late stages of infection. Surprisingly, vgRNA accumulates into distinct globular clusters in the cytoplasmic perinuclear region, which grow and accommodate more vgRNA molecules as infection time increases. The localization of endoplasmic reticulum (ER) markers and nsp3 (a component of the double-membrane vesicle, DMV) at the periphery of the vgRNA clusters suggests that replication organelles are encapsulated into DMVs, which have membranes derived from the host ER. These organelles merge into larger vesicle packets as infection advances. Precise co-imaging of the nanoscale cellular organization of vgRNA, dsRNA, and viral proteins in replication organelles of SARS-CoV-2 may inform therapeutic approaches that target viral replication and associated processes.


Subject(s)
Endoplasmic Reticulum , Organelles , RNA, Viral , SARS-CoV-2 , Virus Replication , SARS-CoV-2/physiology , SARS-CoV-2/ultrastructure , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA, Viral/metabolism , RNA, Viral/genetics , Virus Replication/physiology , Humans , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Endoplasmic Reticulum/ultrastructure , Organelles/virology , Organelles/metabolism , Organelles/ultrastructure , Chlorocebus aethiops , Vero Cells , Animals , COVID-19/virology , COVID-19/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Microscopy, Fluorescence , Viral Replication Compartments/metabolism , RNA, Double-Stranded/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL