Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Molecules ; 27(19)2022 Sep 28.
Article in English | MEDLINE | ID: covidwho-2066278

ABSTRACT

In designing effective siRNAs for a specific mRNA target, it is critically important to have predictive models for the potency of siRNAs. None of the published methods characterized the chemical structures of individual nucleotides constituting a siRNA molecule; therefore, they cannot predict the potency of gene silencing by chemically modified siRNAs (cm-siRNA). We propose a new approach that can predict the potency of gene silencing by cm-siRNAs, which characterizes each nucleotide (NT) using 12 BCUT cheminformatics descriptors describing its charge distribution, hydrophobic and polar properties. Thus, a 21-NT siRNA molecule is described by 252 descriptors resulting from concatenating all the BCUT values of its composing nucleotides. Partial Least Square is employed to develop statistical models. The Huesken data (2431 natural siRNA molecules) were used to perform model building and evaluation for natural siRNAs. Our results were comparable with or superior to those from Huesken's algorithm. The Bramsen dataset (48 cm-siRNAs) was used to build and test the models for cm-siRNAs. The predictive r2 of the resulting models reached 0.65 (or Pearson r values of 0.82). Thus, this new method can be used to successfully model gene silencing potency by both natural and chemically modified siRNA molecules.


Subject(s)
Cheminformatics , Gene Silencing , Nucleotides/genetics , RNA Interference , RNA, Messenger , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
2.
Cell Mol Immunol ; 19(1): 67-78, 2022 01.
Article in English | MEDLINE | ID: covidwho-1541184

ABSTRACT

The global coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused severe morbidity and mortality in humans. It is urgent to understand the function of viral genes. However, the function of open reading frame 10 (ORF10), which is uniquely expressed by SARS-CoV-2, remains unclear. In this study, we showed that overexpression of ORF10 markedly suppressed the expression of type I interferon (IFN-I) genes and IFN-stimulated genes. Then, mitochondrial antiviral signaling protein (MAVS) was identified as the target via which ORF10 suppresses the IFN-I signaling pathway, and MAVS was found to be degraded through the ORF10-induced autophagy pathway. Furthermore, overexpression of ORF10 promoted the accumulation of LC3 in mitochondria and induced mitophagy. Mechanistically, ORF10 was translocated to mitochondria by interacting with the mitophagy receptor Nip3-like protein X (NIX) and induced mitophagy through its interaction with both NIX and LC3B. Moreover, knockdown of NIX expression blocked mitophagy activation, MAVS degradation, and IFN-I signaling pathway inhibition by ORF10. Consistent with our observations, in the context of SARS-CoV-2 infection, ORF10 inhibited MAVS expression and facilitated viral replication. In brief, our results reveal a novel mechanism by which SARS-CoV-2 inhibits the innate immune response; that is, ORF10 induces mitophagy-mediated MAVS degradation by binding to NIX.


Subject(s)
COVID-19/genetics , COVID-19/virology , Immunity, Innate/immunology , Open Reading Frames , SARS-CoV-2/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents/metabolism , Autophagy/immunology , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , Interferon Type I/metabolism , Mitochondria/metabolism , Mitophagy , Proteasome Endopeptidase Complex/metabolism , Ubiquitination , Viral Proteins/metabolism , Virus Replication
4.
Cytokine ; 146: 155634, 2021 10.
Article in English | MEDLINE | ID: covidwho-1293703

ABSTRACT

Thrombopoietin (TPO) is most recognized for its function as the primary regulator of megakaryocyte (MK) expansion and differentiation. MKs, in turn, are best known for their role in platelet production. Research indicates that MKs and platelets play an extensive role in the pathologic thrombosis at sites of high inflammation. TPO, therefore, is a key mediator of thromboinflammation. Silencing of TPO has been shown to decrease platelets levels and rates of pathologic thrombosis in patients with various inflammatory disorders (Barrett et al, 2020; Bunting et al, 1997; Desai et al, 2018; Kaser et al, 2001; Shirai et al, 2019). Given the high rates of thromboinflammmation in the novel coronavirus 2019 (COVID-19), as well as the well-documented aberrant MK activity in affected patients, TPO silencing offers a potential therapeutic modality in the treatment of COVID-19 and other pathologies associated with thromboinflammation. The current review explores the current clinical applications of TPO silencing and offers insight into a potential role in the treatment of COVID-19.


Subject(s)
COVID-19/therapy , Gene Silencing , Inflammation/genetics , Thrombocytosis/genetics , Thrombopoietin/genetics , Thrombosis/genetics , COVID-19/complications , COVID-19/virology , Humans , Inflammation/complications , Inflammation/metabolism , Megakaryocytes/metabolism , SARS-CoV-2/physiology , Thrombocytosis/complications , Thrombocytosis/metabolism , Thrombopoiesis/genetics , Thrombopoietin/metabolism , Thrombosis/complications , Thrombosis/metabolism
5.
Infect Genet Evol ; 93: 104951, 2021 09.
Article in English | MEDLINE | ID: covidwho-1253387

ABSTRACT

The devastating outbreak of COVID-19 has spread all over the world and has become a global health concern. There is no specific therapeutics to encounter the COVID-19. Small interfering RNA (siRNA)-based therapy is an efficient strategy to control human viral infections employing post-transcriptional gene silencing (PTGS) through neutralizing target complementary mRNA. RNA-dependent RNA polymerase (RdRp) encoded by the viral RdRp gene as a part of the replication-transcription complex can be adopted as an acceptable target for controlling SARS-CoV-2 mediated infection. Therefore, in the current study, accessible siRNA designing tools, including significant algorithms and parameters, were rationally used to design the candidate siRNAs against SARS-COV-2 encoded RdRp. The designed siRNA molecules possessed adequate nucleotide-based and other features for potent gene silencing. The targets of the designed siRNAs revealed no significant matches within the whole human genome, ruling out any possibilities for off-target silencing by the siRNAs. Characterization with different potential parameters of efficacy allowed selecting the finest siRNA among all the designed siRNA molecules. Further, validation assessment and target site accessibility prediction also rationalized the suitability of this siRNA molecule. Molecular docking study between the selected siRNA molecule and component of RNA interference (RNAi) pathway gave an excellent outcome. Molecular dynamics of two complexes: siRNA and argonaute complex, guide RNA, and target protein complex, have shown structural stability of these proteins. Therefore, the designed siRNA molecule might act as an effective therapeutic agent against the SARS-CoV-2 at the genome level and can prevent further outbreaks of COVID-19 in humans.


Subject(s)
Coronavirus RNA-Dependent RNA Polymerase/genetics , RNA, Small Interfering/genetics , SARS-CoV-2/genetics , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Base Composition , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Gene Silencing , Genome, Human , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/chemistry , Sequence Alignment
6.
Mol Ther ; 29(7): 2219-2226, 2021 07 07.
Article in English | MEDLINE | ID: covidwho-1228174

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans. Despite several emerging vaccines, there remains no verifiable therapeutic targeted specifically to the virus. Here we present a highly effective small interfering RNA (siRNA) therapeutic against SARS-CoV-2 infection using a novel lipid nanoparticle (LNP) delivery system. Multiple siRNAs targeting highly conserved regions of the SARS-CoV-2 virus were screened, and three candidate siRNAs emerged that effectively inhibit the virus by greater than 90% either alone or in combination with one another. We simultaneously developed and screened two novel LNP formulations for the delivery of these candidate siRNA therapeutics to the lungs, an organ that incurs immense damage during SARS-CoV-2 infection. Encapsulation of siRNAs in these LNPs followed by in vivo injection demonstrated robust repression of virus in the lungs and a pronounced survival advantage to the treated mice. Our LNP-siRNA approaches are scalable and can be administered upon the first sign of SARS-CoV-2 infection in humans. We suggest that an siRNA-LNP therapeutic approach could prove highly useful in treating COVID-19 disease as an adjunctive therapy to current vaccine strategies.


Subject(s)
COVID-19 Drug Treatment , Drug Delivery Systems/methods , Lipids/chemistry , Nanoparticles/chemistry , RNA, Double-Stranded/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , SARS-CoV-2/genetics , Administration, Intravenous , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/metabolism , COVID-19/virology , Female , Gene Silencing , HEK293 Cells , Humans , Lung/metabolism , Male , Mice , Mice, Transgenic , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Transcriptome/drug effects , Treatment Outcome
7.
J Med Virol ; 93(4): 2406-2419, 2021 04.
Article in English | MEDLINE | ID: covidwho-1227754

ABSTRACT

The analyses of 2325 severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) genomes revealed 107, 162, and 65 nucleotide substitutions in the coding region of SARS-CoV-2 from the three continents America, Europe, and Asia, respectively. Of these nucleotide substitutions 58, 94, and 37 were nonsynonymous types mostly present in the Nsp2, Nsp3, Spike, and ORF9. A continent-specific phylogram analyses clustered the SARS-CoV-2 in the different group based on the frequency of nucleotide substitutions. Detailed analyses about the continent-specific amino acid changes and their effectiveness by SNAP2 software was investigated. We found 11 common nonsynonymous mutations; among them, two novel effective mutations were identified in ORF9 (S194L and S202N). Intriguingly, ORF9 encodes nucleocapsid phosphoprotein possessing many effective mutations across continents and could be a potential candidate after the spike protein for studying the role of mutation in viral assembly and pathogenesis. Among the two forms of certain frequent mutation, one form is more prevalent in Europe continents (Nsp12:L314, Nsp13:P504, Nsp13:Y541, Spike:G614, and ORF8:L84) while other forms are more prevalent in American (Nsp12:P314, Nsp13:L504, Nsp13:C541, Spike:D614, and ORF8:L84) and Asian continents (Spike:D614), indicating the spatial and temporal dynamics of SARS-CoV-2. We identified highly conserved 38 regions and among these regions, 11 siRNAs were predicted on stringent criteria that can be used to suppress the expression of viral genes and the corresponding reduction of human viral infections. The present investigation provides information on different mutations and will pave the way for differentiating strains based on virulence and their use in the development of better antiviral therapy.


Subject(s)
COVID-19/virology , Mutation , SARS-CoV-2/genetics , Antiviral Agents/pharmacology , Asia/epidemiology , COVID-19/epidemiology , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Papain-Like Proteases/genetics , Europe/epidemiology , Gene Silencing , Genes, Viral , Genome, Viral , Humans , Open Reading Frames , Phosphoproteins/genetics , Phylogeny , RNA, Small Interfering/genetics , SARS-CoV-2/classification , SARS-CoV-2/drug effects , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , COVID-19 Drug Treatment
8.
Cell ; 184(7): 1790-1803.e17, 2021 04 01.
Article in English | MEDLINE | ID: covidwho-1139467

ABSTRACT

The long non-coding RNA (lncRNA) XIST establishes X chromosome inactivation (XCI) in female cells in early development and thereafter is thought to be largely dispensable. Here, we show XIST is continually required in adult human B cells to silence a subset of X-linked immune genes such as TLR7. XIST-dependent genes lack promoter DNA methylation and require continual XIST-dependent histone deacetylation. XIST RNA-directed proteomics and CRISPRi screen reveal distinctive somatic cell-type-specific XIST complexes and identify TRIM28 that mediates Pol II pausing at promoters of X-linked genes in B cells. Single-cell transcriptome data of female patients with either systemic lupus erythematosus or COVID-19 infection revealed XIST dysregulation, reflected by escape of XIST-dependent genes, in CD11c+ atypical memory B cells (ABCs). XIST inactivation with TLR7 agonism suffices to promote isotype-switched ABCs. These results indicate cell-type-specific diversification and function for lncRNA-protein complexes and suggest expanded roles for XIST in sex-differences in biology and medicine.


Subject(s)
B-Lymphocytes/immunology , COVID-19 , Lupus Erythematosus, Systemic , RNA, Long Noncoding/physiology , Toll-Like Receptor 7/immunology , X Chromosome Inactivation , COVID-19/genetics , COVID-19/immunology , Cell Line , DNA Methylation , Female , Gene Silencing , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology
9.
Virus Res ; 294: 198275, 2021 03.
Article in English | MEDLINE | ID: covidwho-997584

ABSTRACT

Tmprss2 is an emerging molecular target which guides cellular infections of SARS-CoV-2, has been earmarked for interventions against the viral pathologies. The study aims to computationally screen and identifies potential miRNAs, following in vitro experimental validation of miRNA-mediated suppression of Tmprss2 for early prevention of COVID-19. Pool of 163 miRNAs, scrutinized for Tmprss2 binding with three miRNA prediction algorithms, ensued 11 common miRNAs. Further, computational negative energies for association, corroborated miRNA-Tmprss2 interactions, whereas three miRNAs (hsa-miR-214, hsa-miR-98 and hsa-miR-32) based on probability scores ≥0.8 and accessibility to Tmprss2 target have been selected in the Sfold tool. Transfection of miRNA(s) in the Caco-2 cells, quantitatively estimated differential expression, confirming silencing of Tmprss2 with maximum gene suppression by hsa-miR-32 employing novel promising role in CoV-2 pathogenesis. The exalted binding of miRNAs to Tmprss2 and suppression of later advocates their utility as molecular tools for prevention of SARS-CoV-2 viral transmission and replication in humans.


Subject(s)
MicroRNAs/metabolism , SARS-CoV-2/physiology , Serine Endopeptidases/genetics , Virus Internalization , Caco-2 Cells , Computational Biology , Computer Simulation , Gene Silencing , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , Nucleic Acid Conformation
10.
J Adv Res ; 30: 133-145, 2021 05.
Article in English | MEDLINE | ID: covidwho-950741

ABSTRACT

Background: Micro-RNAs (miRNAS) are non-coding, small RNAs that have essential roles in different biological processes through silencing genes, they consist of 18-24 nucleotide length RNA molecules. Recently, miRNAs have been viewed as important modulators of viral infections they can function as suppressors of gene expression by targeting cellular or viral RNAs during infection. Aim of review: We describe the biological roles and effects of miRNAs on SARS-CoV-2 life-cycle and pathogenicity, and we discuss the modulation of the immune system with micro-RNAs which would serve as a new foundation for the treatment of SARS-CoV-2 and other viral infections. Key scientific concepts of review: miRNAs are the key players that regulate the expression of the gene in the post-transcriptional phase and have important effects on viral infections, thus are potential targets in the development of novel therapeutics for the treatment of viral infections. Besides, micro-RNAs (miRNAs) modulation of immune-pathogenesis responses to viral infection is one of the most-known indirect effects, which leads to suppressing of the interferon (IFN-α/ß) signalling cascade or upregulation of the IFN-α/ß production another IFN-stimulated gene (ISGs) that inhibit replication of the virus. These virus-mediated alterations in miRNA levels lead to an environment that might either enhance or inhibit virus replication.


Subject(s)
COVID-19/immunology , Immunity/genetics , MicroRNAs/immunology , RNA, Viral/immunology , SARS-CoV-2/genetics , Gene Silencing/immunology , Humans , Interferons/immunology , Signal Transduction/immunology , Up-Regulation/immunology , Virus Diseases/immunology , Virus Replication/immunology
12.
Genes (Basel) ; 11(11)2020 11 16.
Article in English | MEDLINE | ID: covidwho-927185

ABSTRACT

Rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for coronavirus disease 2019 (COVID-19), has led to a global pandemic, failures of local health care systems, and global economic recession. MicroRNAs (miRNAs) have recently emerged as important regulators of viral pathogenesis, particularly among RNA viruses, but the impact of host miRNAs on SARS-CoV-2 infectivity remains unknown. In this study, we utilize the combination of powerful bioinformatic prediction algorithms and miRNA profiling to predict endogenous host miRNAs that may play important roles in regulating SARS-CoV-2 infectivity. We provide a collection of high-probability miRNA binding sites within the SARS-CoV-2 genome as well as within mRNA transcripts of critical viral entry proteins ACE2 and TMPRSS2 and their upstream modulators, the interferons (IFN). By utilizing miRNA profiling datasets of SARS-CoV-2-resistant and -susceptible cell lines, we verify the biological plausibility of the predicted miRNA-target RNA interactions. Finally, we utilize miRNA profiling of SARS-CoV-2-infected cells to identify predicted miRNAs that are differentially regulated in infected cells. In particular, we identify predicted miRNA binders to SARS-CoV-2 ORFs (miR-23a (1ab), miR-29a, -29c (1ab, N), miR-151a, -151b (S), miR-4707-3p (S), miR-298 (5'-UTR), miR-7851-3p (5'-UTR), miR-8075 (5'-UTR)), ACE2 3'-UTR (miR-9-5p, miR-218-5p), TMPRSS2 3'-UTR (let-7d-5p, -7e-5p, miR-494-3p, miR-382-3p, miR-181c-5p), and IFN-α 3'-UTR (miR-361-5p, miR-410-3p). Overall, this study provides insight into potential novel regulatory mechanisms of SARS-CoV-2 by host miRNAs and lays the foundation for future investigation of these miRNAs as potential therapeutic targets or biomarkers.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Genome, Viral , Interferons/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , SARS-CoV-2/genetics , Serine Endopeptidases/genetics , Angiotensin-Converting Enzyme 2/metabolism , Computational Biology/methods , Gene Silencing , Humans , Interferons/metabolism , MicroRNAs/chemistry , MicroRNAs/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Serine Endopeptidases/metabolism , Transcriptome , Viral Proteins/genetics
13.
Viruses ; 12(9)2020 08 23.
Article in English | MEDLINE | ID: covidwho-727450

ABSTRACT

RNA interference (RNAi) provides the means for alternative antiviral therapy. Delivery of RNAi in the form of short interfering RNA (siRNA), short hairpin RNA (shRNA) and micro-RNA (miRNA) have demonstrated efficacy in gene silencing for therapeutic applications against viral diseases. Bioinformatics has played an important role in the design of efficient RNAi sequences targeting various pathogenic viruses. However, stability and delivery of RNAi molecules have presented serious obstacles for reaching therapeutic efficacy. For this reason, RNA modifications and formulation of nanoparticles have proven useful for non-viral delivery of RNAi molecules. On the other hand, utilization of viral vectors and particularly self-replicating RNA virus vectors can be considered as an attractive alternative. In this review, examples of antiviral therapy applying RNAi-based approaches in various animal models will be described. Due to the current coronavirus pandemic, a special emphasis will be dedicated to targeting Coronavirus Disease-19 (COVID-19).


Subject(s)
Coronavirus Infections/therapy , Pneumonia, Viral/therapy , RNA Interference , Animals , Antiviral Agents/therapeutic use , COVID-19 , Computational Biology , Gene Silencing , Humans , Immunocompromised Host , Pandemics
14.
Front Cell Infect Microbiol ; 10: 290, 2020.
Article in English | MEDLINE | ID: covidwho-612813

ABSTRACT

The recent pandemic SARS-CoV-2 outbreak affects all kinds of individuals worldwide. The health, social, and economic impacts of the pandemic are dramatic, and vaccines or specific treatment options are not yet available. The only approaches that we currently have available to stop the epidemic are those of classical epidemic control, such as case isolation, contact tracing and quarantine, physical distancing, and hygiene measures. It is therefore essential to find further preventive measures and possible interventions that can slow down the number of infected individuals and decrease the severity of disease when affected by SARS-CoV-2. It seems that epigenetic mechanisms are an important part of the pathophysiology and illness severity of COVID-19. These mechanisms have been identified in SARS-CoV-2 but also in other viral infections. If and when these mechanisms are confirmed, then epigenetic interventions influencing DNA methylation could be indicated as primary and/or secondary preventive options.


Subject(s)
Betacoronavirus/genetics , Betacoronavirus/physiology , Coronavirus Infections/virology , DNA Methylation , Epigenesis, Genetic , Lung/virology , Pneumonia, Viral/virology , Virus Internalization , Aging , Angiotensin-Converting Enzyme 2 , COVID-19 , Cell Fusion , Coronavirus Infections/drug therapy , Disease Susceptibility , Female , Gene Silencing , Humans , Male , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/drug therapy , Receptors, Coronavirus , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2 , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL