Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Viruses ; 15(5)2023 05 16.
Article in English | MEDLINE | ID: covidwho-20235842

ABSTRACT

miRNAs, small non-coding RNAs that regulate gene expression, are involved in various pathological processes, including viral infections. Virus infections may interfere with the miRNA pathway through the inhibition of genes involved in miRNA biogenesis. A reduction in the number and the levels of miRNAs expressed in nasopharyngeal swabs of patients with severe COVID-19 was lately observed by us, pointing towards the potential of miRNAs as possible diagnostic or prognostic biomarkers for predicting outcomes among patients with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. The objective of the present study was to investigate whether SARS-CoV-2 infection influences the expression levels of messenger RNAs (mRNAs) of key genes involved in miRNA biogenesis. mRNA levels of AGO2, DICER1, DGCR8, DROSHA, and Exportin-5 (XPO5) were measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR) in nasopharyngeal swab specimens from patients with COVID-19 and controls, as well as in cells infected with SARS-CoV-2 in vitro. Our data showed that the mRNA expression levels of AGO2, DICER1, DGCR8, DROSHA, and XPO5 were not significantly different in patients with severe COVID-19 when compared to patients with non-severe COVID-19 and controls. Similarly, the mRNA expression of these genes was not affected by SARS-CoV-2 infection in NHBE and Calu-3 cells. However, in Vero E6 cells, AGO2, DICER1, DGCR8, and XPO5 mRNA levels were slightly upregulated 24 h after infection with SARS-CoV-2. In conclusion, we did not find evidence for downregulation of mRNA levels of miRNA biogenesis genes during SARS-CoV-2 infection, neither ex vivo nor in vitro.


Subject(s)
COVID-19 , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , COVID-19/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA-Binding Proteins/metabolism , RNA, Messenger/genetics , Ribonuclease III/genetics , Ribonuclease III/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Karyopherins/genetics
2.
Viruses ; 14(10)2022 10 16.
Article in English | MEDLINE | ID: covidwho-2071840

ABSTRACT

Host-virus protein interactions are critical for intracellular viral propagation. Understanding the interactions between cellular and viral proteins may help us develop new antiviral strategies. Porcine epidemic diarrhea virus (PEDV) is a highly contagious coronavirus that causes severe damage to the global swine industry. Here, we employed co-immunoprecipitation and liquid chromatography-mass spectrometry to characterize 426 unique PEDV nucleocapsid (N) protein-binding proteins in infected Vero cells. A protein-protein interaction network (PPI) was created, and gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) database analyses revealed that the PEDV N-bound proteins belong to different cellular pathways, such as nucleic acid binding, ribonucleoprotein complex binding, RNA methyltransferase, and polymerase activities. Interactions of the PEDV N protein with 11 putative proteins: tripartite motif containing 21, DEAD-box RNA helicase 24, G3BP stress granule assembly factor 1, heat shock protein family A member 8, heat shock protein 90 alpha family class B member 1, YTH domain containing 1, nucleolin, Y-box binding protein 1, vimentin, heterogeneous nuclear ribonucleoprotein A2/B1, and karyopherin subunit alpha 1, were further confirmed by in vitro co-immunoprecipitation assay. In summary, studying an interaction network can facilitate the identification of antiviral therapeutic strategies and novel targets for PEDV infection.


Subject(s)
Coronavirus Infections , Nucleic Acids , Porcine epidemic diarrhea virus , Swine Diseases , Chlorocebus aethiops , Swine , Animals , Porcine epidemic diarrhea virus/genetics , Vimentin/metabolism , Vero Cells , Nucleocapsid/metabolism , Nucleocapsid Proteins/genetics , Viral Proteins/metabolism , Coronavirus Infections/metabolism , Antiviral Agents/metabolism , RNA/metabolism , Heat-Shock Proteins/metabolism , Methyltransferases/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , DEAD-box RNA Helicases/metabolism , Ribonucleoproteins/metabolism , Karyopherins/metabolism , Nucleic Acids/metabolism
3.
EMBO Rep ; 23(11): e54061, 2022 Nov 07.
Article in English | MEDLINE | ID: covidwho-2056517

ABSTRACT

Genome-wide screens are powerful approaches to unravel regulators of viral infections. Here, a CRISPR screen identifies the RNA helicase DDX42 as an intrinsic antiviral inhibitor of HIV-1. Depletion of endogenous DDX42 increases HIV-1 DNA accumulation and infection in cell lines and primary cells. DDX42 overexpression inhibits HIV-1 infection, whereas expression of a dominant-negative mutant increases infection. Importantly, DDX42 also restricts LINE-1 retrotransposition and infection with other retroviruses and positive-strand RNA viruses, including CHIKV and SARS-CoV-2. However, DDX42 does not impact the replication of several negative-strand RNA viruses, arguing against an unspecific effect on target cells, which is confirmed by RNA-seq analysis. Proximity ligation assays show DDX42 in the vicinity of viral elements, and cross-linking RNA immunoprecipitation confirms a specific interaction of DDX42 with RNAs from sensitive viruses. Moreover, recombinant DDX42 inhibits HIV-1 reverse transcription in vitro. Together, our data strongly suggest a direct mode of action of DDX42 on viral ribonucleoprotein complexes. Our results identify DDX42 as an intrinsic viral inhibitor, opening new perspectives to target the life cycle of numerous RNA viruses.


Subject(s)
DEAD-box RNA Helicases , HIV-1 , Positive-Strand RNA Viruses , Virus Replication , Humans , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , HIV-1/physiology , Positive-Strand RNA Viruses/physiology , SARS-CoV-2/physiology
4.
Viruses ; 14(10)2022 09 21.
Article in English | MEDLINE | ID: covidwho-2043983

ABSTRACT

In this study, we developed a novel, multiplex qPCR assay for simultaneous detection of RIG-1, MDA5, and IFIT-1 at the mRNA level. The assay was validated in A549 cells transfected with in vitro transcribed RNAs. Both exogenous RNA-GFP and self-amplifying (saRNA-GFP) induced significant expression of RIG-1, MDA5, IFIT-1, as well as type I and III interferons. In contrast, native RNA from intact A549 cells did not upregulate expression of these genes. Next, we evaluated RIG-1, MDA5, and IFIT-1 mRNA levels in the white blood cells of patients with influenza A virus (H3N2) or SARS-CoV-2. In acute phase (about 4 days after disease onset) both viruses induced these genes expression. Clinical observations of SARS-CoV-2 typically describe a two-step disease progression, starting with a mild-to-moderate presentation followed by a secondary respiratory worsening 9 to 12 days after the first onset of symptoms. It revealed that the expression of RIG-1, MDA5, and MxA was not increased after 2 and 3 weeks from the onset the disease, while for IFIT-1 it was observed the second peak at 21 day post infection. It is well known that RIG-1, MDA5, and IFIT-1 expression is induced by the action of interferons. Due to the ability of SOCS-1 to inhibit interferon-dependent signaling, and the distinct antagonism of SARS-CoV-2 in relation to interferon-stimulated genes expression, we assessed SOCS-1 mRNA levels in white blood cells. SARS-CoV-2 patients had increased SOCS-1 expression, while the influenza-infected group did not differ from heathy donors. Moreover, SOCS-1 mRNA expression remained stably elevated during the course of the disease. It can be assumed that augmented SOCS-1 expression is one of multiple mechanisms that allow SARS-CoV-2 to escape from the interferon-mediated immune response. Our results implicate SOCS-1 involvement in the pathogenesis of SARS-CoV-2.


Subject(s)
COVID-19 , Interferons , Humans , Interferons/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Influenza A Virus, H3N2 Subtype/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , SARS-CoV-2/genetics , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , RNA-Binding Proteins , RNA, Messenger/genetics , Antiviral Agents
5.
Front Immunol ; 13: 987453, 2022.
Article in English | MEDLINE | ID: covidwho-2039679

ABSTRACT

The innate immune response to viruses is critical for the correct establishment of protective adaptive immunity. Amongst the many pathways involved, the NLRP3 [nucleotide-binding oligomerisation domain (NOD)-like receptor protein 3 (NLRP3)] inflammasome has received considerable attention, particularly in the context of immunity and pathogenesis during infection with influenza A (IAV) and SARS-CoV-2, the causative agent of COVID-19. Activation of the NLRP3 inflammasome results in the secretion of the proinflammatory cytokines IL-1ß and IL-18, commonly coupled with pyroptotic cell death. While this mechanism is protective and key to host defense, aberrant NLRP3 inflammasome activation causes a hyperinflammatory response and excessive release of cytokines, both locally and systemically. Here, we discuss key molecules in the NLRP3 pathway that have also been shown to have significant roles in innate and adaptive immunity to viruses, including DEAD box helicase X-linked (DDX3X), vimentin and macrophage migration inhibitory factor (MIF). We also discuss the clinical opportunities to suppress NLRP3-mediated inflammation and reduce disease severity.


Subject(s)
COVID-19 , Macrophage Migration-Inhibitory Factors , Carrier Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Humans , Inflammasomes/metabolism , Interleukin-18/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleotides/metabolism , SARS-CoV-2 , Vimentin/metabolism
6.
Cells ; 11(10)2022 05 11.
Article in English | MEDLINE | ID: covidwho-1875501

ABSTRACT

DEAD/H-box proteins are the largest family of RNA helicases in mammalian genomes, and they are present in all kingdoms of life. Since their discovery in the late 1980s, DEAD/H-box family proteins have been a major focus of study. They have been found to play central roles in RNA metabolism, gene expression, signal transduction, programmed cell death, and the immune response to bacterial and viral infections. Aberrant functions of DEAD/H-box proteins have been implicated in a wide range of human diseases that include cancer, neurodegeneration, and inherited genetic disorders. In this review, we provide a historical context and discuss the molecular functions of DEAD/H-box proteins, highlighting the recent discoveries linking their dysregulation to human diseases. We will also discuss the state of knowledge regarding two specific DEAD/H-box proteins that have critical roles in immune responses and programmed cell death, DDX3X and DDX58, also known as RIG-I. Given their importance in homeostasis and disease, an improved understanding of DEAD/H-box protein biology and protein-protein interactions will be critical for informing strategies to counteract the pathogenesis associated with several human diseases.


Subject(s)
DEAD-box RNA Helicases , RNA , Animals , Cell Death , Cell Differentiation , DEAD-box RNA Helicases/metabolism , DNA Helicases , Humans , Inflammation , Mammals/metabolism , RNA/metabolism
7.
Sci Adv ; 8(6): eabk2691, 2022 02 11.
Article in English | MEDLINE | ID: covidwho-1685473

ABSTRACT

Upon virus infection, CD8+ T cell accumulation is tightly controlled by simultaneous proliferation and apoptosis. However, it remains unclear how TCR signal coordinates these events to achieve expansion and effector cell differentiation. We found that T cell-specific deletion of nuclear helicase Dhx9 led to impaired CD8+ T cell survival, effector differentiation, and viral clearance. Mechanistically, Dhx9 acts as the key regulator to ensure LCK- and CD3ε-mediated ZAP70 phosphorylation and ERK activation to protect CD8+ T cells from apoptosis before proliferative burst. Dhx9 directly regulates Id2 transcription to control effector CD8+ T cell differentiation. The DSRM and OB_Fold domains are required for LCK binding and Id2 transcription, respectively. Dhx9 expression is predominantly increased in effector CD8+ T cells of COVID-19 patients. Therefore, we revealed a previously unknown regulatory mechanism that Dhx9 protects activated CD8+ T cells from apoptosis and ensures effector differentiation to promote antiviral immunity independent of nuclear sensor function.


Subject(s)
Antiviral Agents/pharmacology , Arenaviridae Infections/prevention & control , CD8-Positive T-Lymphocytes/immunology , COVID-19/prevention & control , DEAD-box RNA Helicases/metabolism , Immunity, Innate , Neoplasm Proteins/metabolism , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/metabolism , Arenaviridae Infections/pathology , COVID-19/immunology , COVID-19/metabolism , COVID-19/pathology , Cell Differentiation , DEAD-box RNA Helicases/genetics , Humans , Lymphocyte Activation , Lymphocytic choriomeningitis virus/physiology , Mice , Neoplasm Proteins/genetics , SARS-CoV-2/physiology , Virus Replication
8.
Elife ; 112022 01 21.
Article in English | MEDLINE | ID: covidwho-1643864

ABSTRACT

Background: Variants in IFIH1, a gene coding the cytoplasmatic RNA sensor MDA5, regulate the response to viral infections. We hypothesized that IFIH1 rs199076 variants would modulate host response and outcome after severe COVID-19. Methods: Patients admitted to an intensive care unit (ICU) with confirmed COVID-19 were prospectively studied and rs1990760 variants determined. Peripheral blood gene expression, cell populations, and immune mediators were measured. Peripheral blood mononuclear cells from healthy volunteers were exposed to an MDA5 agonist and dexamethasone ex-vivo, and changes in gene expression assessed. ICU discharge and hospital death were modeled using rs1990760 variants and dexamethasone as factors in this cohort and in-silico clinical trials. Results: About 227 patients were studied. Patients with the IFIH1 rs1990760 TT variant showed a lower expression of inflammation-related pathways, an anti-inflammatory cell profile, and lower concentrations of pro-inflammatory mediators. Cells with TT variant exposed to an MDA5 agonist showed an increase in IL6 expression after dexamethasone treatment. All patients with the TT variant not treated with steroids survived their ICU stay (hazard ratio [HR]: 2.49, 95% confidence interval [CI]: 1.29-4.79). Patients with a TT variant treated with dexamethasone showed an increased hospital mortality (HR: 2.19, 95% CI: 1.01-4.87) and serum IL-6. In-silico clinical trials supported these findings. Conclusions: COVID-19 patients with the IFIH1 rs1990760 TT variant show an attenuated inflammatory response and better outcomes. Dexamethasone may reverse this anti-inflammatory phenotype. Funding: Centro de Investigación Biomédica en Red (CB17/06/00021), Instituto de Salud Carlos III (PI19/00184 and PI20/01360), and Fundació La Marató de TV3 (413/C/2021).


Patients with severe COVID-19 often need mechanical ventilation to help them breathe and other types of intensive care. The outcome for many of these patients depends on how their immune system reacts to the infection. If the inflammatory response triggered by the immune system is too strong, this can cause further harm to the patient. One gene that plays an important role in inflammation is IFIH1 which encodes a protein that helps the body to recognize viruses. There are multiple versions of this gene which each produce a slightly different protein. It is possible that this variation impacts how the immune system responds to the virus that causes COVID-19. To investigate, Amado-Rodríguez, Salgado del Riego et al. analyzed the IFIH1 gene in 227 patients admitted to an intensive care unit in Spain for severe COVID-19 between March and December 2020. They found that patients with a specific version of the gene called TT experienced less inflammation and were more likely to survive the infection. Physicians typically treat patients with moderate to severe COVID-19 with corticosteroid drugs that reduce the inflammatory response. However, Amado-Rodríguez, Salgado del Riego et al. found that patients with the TT version of the IFIH1 gene were at greater risk of dying if they received corticosteroids. The team then applied the distribution of IFIH1 variants among different ethnic ancestries to data from a previous clinical trial, and simulated the effects of corticosteroid treatment. This 'mock' clinical trial supported their findings from the patient-derived data, which were also validated by laboratory experiments on immune cells from individuals with the TT gene. The work by Amado-Rodríguez, Salgado del Riego et al. suggests that while corticosteroids benefit some patients, they may cause harm to others. However, a real-world clinical trial is needed to determine whether patients with the TT version of the IFIH1 gene would do better without steroids.


Subject(s)
COVID-19/genetics , Inflammation/genetics , Interferon-Induced Helicase, IFIH1/genetics , SARS-CoV-2/pathogenicity , Aged , COVID-19/complications , Critical Illness , DEAD-box RNA Helicases/metabolism , Female , Humans , Inflammation/metabolism , Male , Middle Aged
9.
Science ; 373(6551): 231-236, 2021 07 09.
Article in English | MEDLINE | ID: covidwho-1304152

ABSTRACT

In mammals, early resistance to viruses relies on interferons, which protect differentiated cells but not stem cells from viral replication. Many other organisms rely instead on RNA interference (RNAi) mediated by a specialized Dicer protein that cleaves viral double-stranded RNA. Whether RNAi also contributes to mammalian antiviral immunity remains controversial. We identified an isoform of Dicer, named antiviral Dicer (aviD), that protects tissue stem cells from RNA viruses-including Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-by dicing viral double-stranded RNA to orchestrate antiviral RNAi. Our work sheds light on the molecular regulation of antiviral RNAi in mammalian innate immunity, in which different cell-intrinsic antiviral pathways can be tailored to the differentiation status of cells.


Subject(s)
DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , RNA Interference , RNA Viruses/physiology , RNA, Viral/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Stem Cells/enzymology , Stem Cells/virology , Alternative Splicing , Animals , Brain/enzymology , Brain/virology , Cell Line , DEAD-box RNA Helicases/chemistry , Humans , Immunity, Innate , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Organoids/enzymology , Organoids/virology , RNA Virus Infections/enzymology , RNA Virus Infections/immunology , RNA Virus Infections/virology , RNA Viruses/genetics , RNA Viruses/immunology , RNA, Double-Stranded/metabolism , RNA, Small Interfering/metabolism , Ribonuclease III/chemistry , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Virus Replication , Zika Virus/genetics , Zika Virus/immunology , Zika Virus/physiology , Zika Virus Infection/enzymology , Zika Virus Infection/immunology , Zika Virus Infection/virology
10.
Antiviral Res ; 190: 105064, 2021 06.
Article in English | MEDLINE | ID: covidwho-1157118

ABSTRACT

COVID-19 is currently a highly pressing health threat and therapeutic strategies to mitigate the infection impact are urgently needed. Characterization of the SARS-CoV-2 interactome in infected cells may represent a powerful tool to identify cellular proteins hijacked by viruses for their life cycle and develop host-oriented antiviral therapeutics. Here we report the proteomic characterization of host proteins interacting with SARS-CoV-2 Nucleoprotein in infected Vero E6 cells. We identified 24 high-confidence proteins mainly playing a role in RNA metabolism and translation, including RNA helicases and scaffold proteins involved in the formation of stress granules, cytoplasmic aggregates of messenger ribonucleoproteins that accumulate as a result of stress-induced translation arrest. Analysis of stress granules upon SARS-CoV-2 infection showed that these structures are not induced in infected cells, neither eIF2α phosphorylation, an upstream event leading to stress-induced translation inhibition. Notably, we found that G3BP1, a stress granule component that associates with the Nucleoprotein, is required for efficient SARS-CoV-2 replication. Moreover, we showed that the Nucleoprotein-interacting RNA helicase DDX3X colocalizes with viral RNA foci and its inhibition by small molecules or small interfering RNAs significantly reduces viral replication. Altogether, these results indicate that SARS-CoV-2 subverts the stress granule machinery and exploits G3BP1 and DDX3X for its replication cycle, offering groundwork for future development of host-directed therapies.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , COVID-19/metabolism , DEAD-box RNA Helicases/metabolism , Animals , COVID-19/virology , Cell Line , Chlorocebus aethiops , DNA Helicases , Eukaryotic Initiation Factor-2/metabolism , Host-Pathogen Interactions , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Proteomics/methods , RNA Helicases , RNA Recognition Motif Proteins/metabolism , RNA, Small Interfering/metabolism , RNA, Viral/metabolism , SARS-CoV-2/metabolism , Vero Cells , Virus Replication/physiology
11.
Nucleic Acids Res ; 49(7): e40, 2021 04 19.
Article in English | MEDLINE | ID: covidwho-1050155

ABSTRACT

Generation of conditional knockout (cKO) and various gene-modified cells is laborious and time-consuming. Here, we established an all-in-one cKO system, which enables highly efficient generation of cKO cells and simultaneous gene modifications, including epitope tagging and reporter gene knock-in. We applied this system to mouse embryonic stem cells (ESCs) and generated RNA helicase Ddx1 cKO ESCs. The targeted cells displayed endogenous promoter-driven EGFP and FLAG-tagged DDX1 expression, and they were converted to Ddx1 KO via FLP recombinase. We further established TetFE ESCs, which carried a reverse tetracycline transactivator (rtTA) expression cassette and a tetracycline response element (TRE)-regulated FLPERT2 cassette in the Gt(ROSA26)Sor locus for instant and tightly regulated induction of gene KO. By utilizing TetFE Ddx1F/F ESCs, we isolated highly pure Ddx1F/F and Ddx1-/- ESCs and found that loss of Ddx1 caused rRNA processing defects, thereby activating the ribosome stress-p53 pathway. We also demonstrated cKO of various genes in ESCs and homologous recombination-non-proficient human HT1080 cells. The frequency of cKO clones was remarkably high for both cell types and reached up to 96% when EGFP-positive clones were analyzed. This all-in-one cKO system will be a powerful tool for rapid and precise analyses of gene functions.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Knockout Techniques/methods , RNA, Ribosomal/metabolism , Animals , Cell Line , Embryonic Stem Cells , Fibroblasts , Gene Expression , Gene Knock-In Techniques , Humans , Mice , Mice, Inbred C57BL , RNA Processing, Post-Transcriptional , Ribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL