Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Nature ; 588(7836): 169-173, 2020 12.
Article in English | MEDLINE | ID: mdl-33087935

ABSTRACT

Cancer therapies that target epigenetic repressors can mediate their effects by activating retroelements within the human genome. Retroelement transcripts can form double-stranded RNA (dsRNA) that activates the MDA5 pattern recognition receptor1-6. This state of viral mimicry leads to loss of cancer cell fitness and stimulates innate and adaptive immune responses7,8. However, the clinical efficacy of epigenetic therapies has been limited. To find targets that would synergize with the viral mimicry response, we sought to identify the immunogenic retroelements that are activated by epigenetic therapies. Here we show that intronic and intergenic SINE elements, specifically inverted-repeat Alus, are the major source of drug-induced immunogenic dsRNA. These inverted-repeat Alus are frequently located downstream of 'orphan' CpG islands9. In mammals, the ADAR1 enzyme targets and destabilizes inverted-repeat Alu dsRNA10, which prevents activation of the MDA5 receptor11. We found that ADAR1 establishes a negative-feedback loop, restricting the viral mimicry response to epigenetic therapy. Depletion of ADAR1 in patient-derived cancer cells potentiates the efficacy of epigenetic therapy, restraining tumour growth and reducing cancer initiation. Therefore, epigenetic therapies trigger viral mimicry by inducing a subset of inverted-repeats Alus, leading to an ADAR1 dependency. Our findings suggest that combining epigenetic therapies with ADAR1 inhibitors represents a promising strategy for cancer treatment.


Subject(s)
Adenosine Deaminase/metabolism , Alu Elements/drug effects , Alu Elements/genetics , Decitabine/pharmacology , Decitabine/therapeutic use , Epigenesis, Genetic/drug effects , RNA-Binding Proteins/metabolism , Transcription, Genetic/drug effects , Adaptive Immunity/drug effects , Adenosine Deaminase/deficiency , Alu Elements/immunology , Animals , Cell Line, Tumor , CpG Islands/drug effects , CpG Islands/genetics , DNA, Intergenic/drug effects , DNA, Intergenic/genetics , DNA, Intergenic/immunology , DNA-Cytosine Methylases/antagonists & inhibitors , Feedback, Physiological , Humans , Immunity, Innate/drug effects , Interferon-Induced Helicase, IFIH1/metabolism , Introns/drug effects , Introns/genetics , Introns/immunology , Inverted Repeat Sequences/drug effects , Inverted Repeat Sequences/genetics , Inverted Repeat Sequences/immunology , Male , Mice , Molecular Mimicry/drug effects , Molecular Mimicry/immunology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , RNA, Double-Stranded/drug effects , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA-Binding Proteins/antagonists & inhibitors , Viruses/drug effects , Viruses/immunology
2.
J Biol Chem ; 296: 100264, 2021.
Article in English | MEDLINE | ID: mdl-33837743

ABSTRACT

Recent studies have demonstrated that embryonic stem cells (ESCs) are deficient in expressing type I interferons (IFN), the cytokines that play key roles in antiviral responses. However, the underlying molecular mechanisms and biological implications of this finding are poorly understood. In this study, we developed a synthetic RNA-based assay that can simultaneously assess multiple forms of antiviral responses. Dicer is an enzyme essential for RNA interference (RNAi), which is used as a major antiviral mechanism in invertebrates. RNAi activity is detected in wild-type ESCs but is abolished in Dicer knockout ESCs (D-/-ESCs) as expected. Surprisingly, D-/-ESCs have gained the ability to express IFN, which is otherwise deficient in wild-type ESCs. Furthermore, D-/-ESCs have constitutively active double-stranded RNA (dsRNA)-activated protein kinase (PKR), an enzyme that is also involved in antiviral response. D-/-ESCs show increased sensitivity to the cytotoxicity resulting from RNA transfection. The effects of dsRNA can be partly replicated with a synthetic B2RNA corresponding to the retrotransposon B2 short interspersed nuclear element. B2RNA has secondary structure features of dsRNA and accumulates in D-/-ESCs, suggesting that B2RNA could be a cellular RNA that activates PKR and contributes to the decreased cell proliferation and viability of D-/-ESCs. Treatment of D-/-ESCs with a PKR inhibitor and IFNß-neutralizing antibodies increased cell proliferation rate and cell viability. Based on these findings, we propose that, in ESCs, Dicer acts as a repressor of antiviral responses and plays a key role in the maintenance of proliferation, viability, and pluripotency of ESCs.


Subject(s)
DEAD-box RNA Helicases/genetics , Interferon Type I/genetics , Interferon-gamma/genetics , Mouse Embryonic Stem Cells/drug effects , Ribonuclease III/genetics , eIF-2 Kinase/genetics , Animals , Antiviral Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference/drug effects , RNA, Double-Stranded/drug effects , RNA, Double-Stranded/genetics , Retroelements/genetics , eIF-2 Kinase/antagonists & inhibitors
3.
J Cell Physiol ; 235(4): 3646-3656, 2020 04.
Article in English | MEDLINE | ID: mdl-31559639

ABSTRACT

It is well known that exposure of double-stranded RNA (dsRNA) to intestine immediately induces villus damage with severe diarrhea, which is mediated by toll-like receptor 3 signaling activation. However, the role of intestinal stem cells (ISCs) remains obscure during the pathology. In the present study, polyinosinic-polycytidylic acid (poly[I:C]), mimicking viral dsRNA, was used to establish intestinal damage model. Mice were acutely and chronically exposed to poly(I:C), and ISCs in jejunum were analyzed. The results showed that the height of villus was shorter 48 hr after acute poly(I:C) exposure compared with that of controls, while chronic poly(I:C) treatment increased both villus height and crypt depth in jejunum compared with control animals. The numbers of ISCs in jejunum were significantly increased after acute and chronic poly(I:C) exposure. Poly (I:C)-stimulated ISCs have stronger capacities to differentiate into intestine endocrine cells. Mechanistically, poly(I:C) treatment increased expression of Stat1 and Axin2 in the intestinal crypt, which was along with increased expression of Myc, Bcl2, and ISC proliferation. These findings suggest that dsRNA exposure could induce ISC proliferation to ameliorate dsRNA-induced intestinal injury.


Subject(s)
Intestinal Mucosa/growth & development , Poly I-C/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Axin Protein/genetics , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Jejunum/drug effects , Jejunum/growth & development , Mice , RNA, Double-Stranded/drug effects , STAT1 Transcription Factor/genetics , Signal Transduction , Toll-Like Receptor 3/genetics
4.
Proc Natl Acad Sci U S A ; 113(37): 10238-44, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27573823

ABSTRACT

Vitamin C deficiency is found in patients with cancer and might complicate various therapy paradigms. Here we show how this deficiency may influence the use of DNA methyltransferase inhibitors (DNMTis) for treatment of hematological neoplasias. In vitro, when vitamin C is added at physiological levels to low doses of the DNMTi 5-aza-2'-deoxycytidine (5-aza-CdR), there is a synergistic inhibition of cancer-cell proliferation and increased apoptosis. These effects are associated with enhanced immune signals including increased expression of bidirectionally transcribed endogenous retrovirus (ERV) transcripts, increased cytosolic dsRNA, and activation of an IFN-inducing cellular response. This synergistic effect is likely the result of both passive DNA demethylation by DNMTi and active conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) by ten-eleven translocation (TET) enzymes at LTR regions of ERVs, because vitamin C acts as a cofactor for TET proteins. In addition, TET2 knockout reduces the synergy between the two compounds. Furthermore, we show that many patients with hematological neoplasia are markedly vitamin C deficient. Thus, our data suggest that correction of vitamin C deficiency in patients with hematological and other cancers may improve responses to epigenetic therapy with DNMTis.


Subject(s)
Ascorbic Acid/administration & dosage , Azacitidine/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Hematologic Neoplasms/drug therapy , Apoptosis/drug effects , Ascorbic Acid Deficiency/complications , Ascorbic Acid Deficiency/drug therapy , Ascorbic Acid Deficiency/metabolism , Ascorbic Acid Deficiency/pathology , Azacitidine/administration & dosage , Cell Proliferation/drug effects , DNA Methylation/drug effects , DNA-Binding Proteins/genetics , Decitabine , Dioxygenases , Drug Synergism , Endogenous Retroviruses/genetics , Female , Hematologic Neoplasms/complications , Hematologic Neoplasms/pathology , Humans , Interferons/genetics , Male , Methyltransferases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Double-Stranded/drug effects
5.
Hepatology ; 66(2): 357-370, 2017 08.
Article in English | MEDLINE | ID: mdl-27642141

ABSTRACT

Hepatitis C virus (HCV) is unique among RNA viruses in its ability to establish chronic infection in the majority of exposed adults. HCV persists in the liver despite interferon (IFN)-stimulated gene (ISG) induction; robust induction actually predicts treatment failure and viral persistence. It is unclear which forms of HCV RNA are associated with ISG induction and IFN resistance during natural infections. To thoroughly delineate HCV RNA populations, we developed conditions that fully separate the strands of long double-stranded RNA (dsRNA) and allow the released RNAs to be quantified in reverse transcription/polymerase chain reaction assays. These methods revealed that dsRNA, a pathogen-associated molecular pattern (PAMP), comprised 52% (standard deviation, 28%) of the HCV RNA in the livers of patients with chronic infection. HCV dsRNA was proportionally higher in patients with the unfavorable IL28B TT (rs12979860) genotype. Higher ratios of HCV double-stranded to single-stranded RNA (ssRNA) correlated positively with ISG induction. In Huh-7.5 cells, IFN treatment increased the total amount of HCV dsRNA through a process that required de novo viral RNA synthesis and shifted the ratio of viral dsRNA/ssRNA in favor of dsRNA. This shift was blocked by ribavirin (RBV), an antiviral drug that reduces relapse in HCV patients. Northern blotting established that HCV dsRNA contained genome-length minus strands. CONCLUSION: HCV dsRNA is the predominant form in the HCV-infected liver and has features of both a PAMP and a genomic reservoir. Interferon treatment increased rather than decreased HCV dsRNA. This unexpected finding suggests that HCV produces dsRNA in response to IFN, potentially to antagonize antiviral defenses. (Hepatology 2017;66:357-370).


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/genetics , Hepatitis C/pathology , Interferon-alpha/pharmacology , RNA, Double-Stranded/genetics , Adult , Biopsy, Needle , Blotting, Western , Cells, Cultured , Female , Flow Cytometry , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/genetics , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Immunohistochemistry , Male , RNA, Double-Stranded/drug effects , RNA, Viral/drug effects , RNA, Viral/genetics , Reference Values , Sensitivity and Specificity
6.
J Virol ; 88(1): 456-68, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24155404

ABSTRACT

Measles virus (MV) lacking expression of C protein (C(KO)) is a potent activator of the double-stranded RNA (dsRNA)-dependent protein kinase (PKR), whereas the isogenic parental virus expressing C protein is not. Here, we demonstrate that significant amounts of dsRNA accumulate during C(KO) mutant infection but not following parental virus infection. dsRNA accumulated during late stages of infection and localized with virus replication sites containing N and P proteins. PKR autophosphorylation and stress granule formation correlated with the timing of dsRNA appearance. Phospho-PKR localized to dsRNA-containing structures as revealed by immunofluorescence. Production of dsRNA was sensitive to cycloheximide but resistant to actinomycin D, suggesting that dsRNA is a viral product. Quantitative PCR (qPCR) analyses revealed reduced viral RNA synthesis and a steepened transcription gradient in C(KO) virus-infected cells compared to those in parental virus-infected cells. The observed alterations were further reflected in lower viral protein expression levels and reduced C(KO) virus infectious yield. RNA deep sequencing confirmed the viral RNA expression profile differences seen by qPCR between C(KO) mutant and parental viruses. After one subsequent passage of the C(KO) virus, defective interfering RNA (DI-RNA) with a duplex structure was obtained that was not seen with the parental virus. We conclude that in the absence of C protein, the amount of PKR activator RNA, including DI-RNA, is increased, thereby triggering innate immune responses leading to impaired MV growth.


Subject(s)
Measles virus/metabolism , Protein Kinases/metabolism , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Viral Proteins/physiology , Base Sequence , Cell Line , Cycloheximide/pharmacology , DNA Primers , Dactinomycin/pharmacology , Enzyme Activation , High-Throughput Nucleotide Sequencing , Humans , Measles virus/genetics , Phosphorylation , Polymerase Chain Reaction , RNA, Double-Stranded/drug effects , RNA, Viral/drug effects
7.
Nucleic Acids Res ; 41(6): 3819-32, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23393189

ABSTRACT

The serotonin receptor 2C plays a central role in mood and appetite control. It undergoes pre-mRNA editing as well as alternative splicing. The RNA editing suggests that the pre-mRNA forms a stable secondary structure in vivo. To identify substances that promote alternative exons inclusion, we set up a high-throughput screen and identified pyrvinium pamoate as a drug-promoting exon inclusion without editing. Circular dichroism spectroscopy indicates that pyrvinium pamoate binds directly to the pre-mRNA and changes its structure. SHAPE (selective 2'-hydroxyl acylation analysed by primer extension) assays show that part of the regulated 5'-splice site forms intramolecular base pairs that are removed by this structural change, which likely allows splice site recognition and exon inclusion. Genome-wide analyses show that pyrvinium pamoate regulates >300 alternative exons that form secondary structures enriched in A-U base pairs. Our data demonstrate that alternative splicing of structured pre-mRNAs can be regulated by small molecules that directly bind to the RNA, which is reminiscent to an RNA riboswitch.


Subject(s)
Alternative Splicing/drug effects , Pyrvinium Compounds/pharmacology , RNA, Messenger/drug effects , Receptor, Serotonin, 5-HT2C/genetics , Base Sequence , Exons , HEK293 Cells , High-Throughput Screening Assays , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Phylogeny , Pyrvinium Compounds/metabolism , RNA Editing , RNA Precursors/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/drug effects , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Spliceosomes/metabolism
8.
Angew Chem Int Ed Engl ; 53(38): 10119-23, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25044924

ABSTRACT

Luminescent metallo-intercalators are potent biosensors of nucleic acid structure and anticancer agents targeting DNAs. There are few examples of luminescent metallo-intercalators which can simultaneously act as emission probes of nucleic acid structure and display promising anticancer activities. Herein, we describe a luminescent platinum(II) complex, [Pt(C^N^N)(C≡NtBu)]ClO4 (1 a, HC^N^N= 6-phenyl-2,2'-bipyridyl), that intercalates between the nucleobases of nucleic acids, accompanied by an increase in emission intensity and/or a significant change in the maximum emission wavelength. The changes in emission properties measured with double-stranded RNA (dsRNA) are different from those with dsDNA used in the binding reactions. Complex 1 a exhibited potent anticancer activity towards cancer cells in vitro and inhibited tumor growth in a mouse model. The stabilization of the topoisomerase I-DNA complex with resulting DNA damage by 1 a is suggested to contribute to its anticancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/drug effects , Luminescence , Neoplasms, Experimental/drug therapy , Organoplatinum Compounds/pharmacology , RNA, Double-Stranded/drug effects , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/pathology , Organoplatinum Compounds/chemistry , RNA, Double-Stranded/chemistry , Structure-Activity Relationship
9.
FASEB J ; 26(8): 3188-98, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22532440

ABSTRACT

Ovarian cancer is the most lethal gynecological cancer. Here we show that innate immune agonist, dsRNA, directly induces ovarian cancer cell death and identify biomarkers associated with responsiveness to this targeted treatment. Nuclear staining and MTT assays following dsRNA stimulation revealed two subpopulations, sensitive (OVCAR-3, CAOV-3; patient samples malignant 1 and 2) and resistant (DOV-13, SKOV-3). Microarray analysis identified 75 genes with differential expression that further delineated these two subpopulations. qPCR and immunoblot analyses showed increased dsRNA receptor expression after stimulation as compared to resistant and immortalized ovarian surface epithelial cells (e.g., 70-fold with malignant 2, 43-fold with OVCAR-3). Using agonists, antagonists, and shRNA-mediated knockdown of dsRNA receptors, we show that TLR3, RIG-I, and mda5 coordinated a caspase 8/9- and interferon-dependent cell death. In resistant cells, dsRNA receptor overexpression restored dsRNA sensitivity. When dsRNA was combined with carboplatin or paclitaxel, cell viability significantly decreased over individual treatments (1.5- to 7.5-fold). Isobologram analyses showed synergism in dsRNA combinations (CI=0.4-0.82) vs. an additive effect in carboplatin/paclitaxel treatment (CI=1.5-2). Our data identify a predictive marker, dsRNA receptor expression, to target dsRNA responsive populations and show that, in dsRNA-sensitive cells, dsRNA induces apoptosis and enhances the potency of cytotoxic chemotherapeutics.


Subject(s)
Apoptosis/drug effects , Ovarian Neoplasms/drug therapy , RNA, Double-Stranded/therapeutic use , Biomarkers, Tumor/analysis , Caspases/metabolism , Cell Line, Tumor , Cisplatin/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Activation , Female , Humans , Interferon-beta/metabolism , NF-kappa B/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , RNA, Double-Stranded/drug effects , RNA, Messenger/metabolism
10.
Pharmazie ; 68(11): 899-903, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24380240

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is elevated in asthma and triggers dendritic cell-mediated activation of TH2 inflammatory responses. Viral stimuli, a major cause of asthma exacerbations, have been shown to induce overexpression of TSLP in asthmatic epithelium. Azithromycin has various anti-microbial and antiinflammatory effects. However, the effect of azithromycin on the production of TSLP has not been studied. Here we explored the effects of azithromycin on viral surrogate (dsRNA)-induced TSLP in normal human bronchial epithelial (NHBE) cells. NHBE were stimulated with poly (I:C) in the presence azithromycin. The effects of azithromycin on dsRNA-induced inflammatory responses in NHBE cells were analyzed. We demonstrated that azithromycin inhibited the production and mRNA expression of TSLP in NHBE cells. Azithromycin also inhibited the nuclear factor-KB luciferase activity induced by poly (I:C), and it prevented dsRNA-induced loss of the NF-kappaB repressor protein IkappaBalpha. These results suggest that azithromycin can be useful to treat asthma exacerbations due to the inhibition of TSLP.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Azithromycin/pharmacology , Cytokines/metabolism , Epithelial Cells/metabolism , RNA, Double-Stranded/drug effects , Respiratory Mucosa/metabolism , Thymus Gland/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Humans , Indicators and Reagents , Interferon-beta/biosynthesis , Interleukin-8/biosynthesis , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Thymus Gland/drug effects , Transfection , Thymic Stromal Lymphopoietin
11.
Mol Immunol ; 129: 32-38, 2021 01.
Article in English | MEDLINE | ID: mdl-33260038

ABSTRACT

Tryptanthrin is a bioactive component of indigo plants such as Polygonum tinctrorium and known to have an anti-inflammatory activity. The aim of this study was to investigate the effects of tryptanthrin on Toll-like receptor 3 (TLR3)-mediated cytokine and chemokine expression in human umbilical vein endothelial cells (HUVEC). Herein, we found that tryptanthrin suppressed the expression of CXCL10 in HUVEC upon stimulation with a TLR3 ligand polyinosinic-polycytidylic acid (poly IC). Tryptanthrin did not inhibit poly IC-induced activation of interferon regulatory factor 3 (IRF3) or the mRNA expression of interferon (IFN)-ß, while it significantly suppressed the expression of RIG-I, MDA5, and classical IFN-stimulated genes (ISGs). Tryptanthrin attenuated the phosphorylation and nuclear translocation of STAT1 in HUVEC stimulated with not only poly IC but also recombinant IFN-ß. These results suggested that tryptanthrin inhibited poly IC-induced expression of CXCL10 and ISGs via suppressing the activation of STAT1 in HUVEC. Our findings indicate that tryptanthrin may be useful for regulating TLR3-mediated vascular inflammation.


Subject(s)
Chemokine CXCL10/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Phosphorylation/drug effects , Quinazolines/pharmacology , RNA, Double-Stranded/drug effects , STAT1 Transcription Factor/metabolism , Cells, Cultured , DEAD Box Protein 58/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-beta/metabolism , Poly I-C/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 3/metabolism
12.
Minerva Med ; 112(1): 144-152, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33104300

ABSTRACT

The world is now entering its 9th month of combat against a pandemic of deadly pneumonia. Started out from China in December 2019, the disease has been declared as caused by infection with a so far unknown RNA Coronavirus of the respiratory family, then named severe acute respiratory syndrome coronavirus SARS-CoV-2. In the absence of a vaccine, and with scientists still struggling for an effective therapy, COVID-19 (the SARS-dependent syndrome) carries up to now, a death toll of more than 590,000 (July 18,2020) undermining jobs and finance of contemporary society in all continents. Social distancing, the only measure hitherto shown to restrain virus spread, has been progressively loosened from May 2020 in some countries, leaving us in the fear of repeat attacks from the unchecked virus. We discuss the problem and propose to tentatively boost the antivirus cell machinery by using lab-made viral mimics to engage cell receptors.


Subject(s)
COVID-19/therapy , SARS-CoV-2 , COVID-19/complications , COVID-19/epidemiology , Carboxymethylcellulose Sodium/analogs & derivatives , Carboxymethylcellulose Sodium/therapeutic use , Cytokine Release Syndrome/etiology , Humans , Immunization, Passive , Interferon Inducers/therapeutic use , Mucocutaneous Lymph Node Syndrome/etiology , Physical Distancing , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Polylysine/therapeutic use , Practice Guidelines as Topic , RNA, Double-Stranded/drug effects , RNA, Viral/drug effects , Recurrence , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Secondary Prevention , COVID-19 Drug Treatment , COVID-19 Serotherapy
13.
Anticancer Res ; 29(4): 1067-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19414347

ABSTRACT

Onconase is a cytotoxic ribonuclease which targets tumor cells in vivo and in vitro. To date, cellular tRNA appeared to be the major target for Onconase mediated cytotoxic activity. Most recently we demonstrated that Onconase can also cleave double-stranded RNA (dsRNA). Incubation of Onconase at 37 degrees C with GAPDH gene-dsRNA (approximately 440 bp long) and dsRNA ladder showed degradation of dsRNA into a spectrum of smaller dsRNA fragments. Moreover, incubation of dsRNA substrates at 40 degrees C under similar conditions markedly potentiated further cleavage of dsRNAs. The recently discovered double-stranded RNase activity of Onconase suggests another mechanism for inducing cell death/apoptosis in malignant phenotypes via the RNA interference mechanism involving siRNA and miRNA.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphocytes/drug effects , RNA, Double-Stranded/drug effects , Ribonucleases/pharmacology , Apoptosis/drug effects , Cells, Cultured , Endoribonucleases/metabolism , Gene Silencing , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , In Vitro Techniques , Lymphocytes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Double-Stranded/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology
14.
Nucleic Acid Ther ; 29(4): 195-207, 2019 08.
Article in English | MEDLINE | ID: mdl-31140918

ABSTRACT

Nanoparticle based siRNA formulations often suffer from aggregation and loss of function during storage. We in this study report a frozen targeted RGD-polyethylene glycol (PEG)-ECO/siß3 nanoparticle formulation with a prolonged shelf life and preserved nanoparticle functionality. The targeted RGD-PEG-ECO/siß3 nanoparticles are formed by step-wised self-assembly of RGD-PEG-maleimide, ECO, and siRNA. The nanoparticles have a diameter of 224.5 ± 9.41 nm and a zeta potential to 45.96 ± 3.67 mV in water and a size of 234.34 ± 3.01 nm and a near neutral zeta potential in saline solution. The addition of sucrose does not affect their size and zeta potential and substantially preserves the integrity and biological activities of frozen and lyophilized formulations of the targeted nanoparticles. The frozen formulation with as low as 5% sucrose retains nanoparticle integrity (90% siRNA encapsulation), size distribution (polydispersity index [PDI] ≤20%), and functionality (at least 75% silencing efficiency) at -80°C for at least 1 year. The frozen RGD-PEG-ECO/siß3 nanoparticle formulation exhibits excellent biocompatibility, with no adverse effects on hemocompatibility and minimal immunogenicity. As RNAi holds the promise in treating the previously untreatable diseases, the frozen nanoparticle formulation with the low sucrose concentration has the potential to be a delivery platform for clinical translation of RNAi therapeutics.


Subject(s)
Biocompatible Materials/pharmacology , Drug Delivery Systems , Nanoparticles/chemistry , RNA, Small Interfering/pharmacology , Biocompatible Materials/chemistry , Cell Line, Tumor , Humans , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , RNA Interference , RNA, Double-Stranded/drug effects , RNA, Double-Stranded/genetics , RNA, Small Interfering/chemistry
15.
Nucleic Acid Ther ; 29(1): 33-43, 2019 02.
Article in English | MEDLINE | ID: mdl-30562146

ABSTRACT

The objective of this study was to analyze the effects of fluorophores on the intracellular accumulation and biological activity of small interfering RNA (siRNA) and its cholesterol conjugates. In this study, we used stem-loop real-time PCR and calibration curves to quantitate cellular siRNA accumulation. Attachment of fluorophores significantly affected both the accumulation and biological activity of siRNA conjugates. The severity of this effect depended significantly on the structure of the conjugate; fluorophores (Cy5.5 or Alexa-488) attached to siRNA, facing the side of the duplex opposite to cholesterol, enhanced the unproductive intracellular accumulation of the conjugate when delivered in carrier-free mode. Enhanced cellular accumulation of siRNA conjugates did not result in enhanced biological activity of the conjugate. Moreover, the attachment of a hydrophobic fluorophore, such as Cy5.5, to conventional siRNA also enhanced its apparent intracellular accumulation, but not its biological activity. Thus, the use of fluorescent labels for the study of the intracellular accumulation of siRNA and its conjugates formed with different molecules is possible only for a limited range of structures, and requires verification using alternative methods.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Cholesterol/pharmacology , RNA, Small Interfering/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Carbocyanines/pharmacology , Cell Line , Cholesterol/chemistry , Drug Resistance/drug effects , Drug Resistance/genetics , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Gene Silencing/drug effects , Humans , RNA Interference/drug effects , RNA, Double-Stranded/drug effects , RNA, Double-Stranded/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
16.
Mol Immunol ; 99: 19-29, 2018 07.
Article in English | MEDLINE | ID: mdl-29674236

ABSTRACT

Ultraviolet B (UVB) irradiation exerts multiple effects on skin cells, inducing apoptosis, senescence and carcinogenesis. Toll-like receptor 3, a member of pattern recognition receptors, is reported to initiate inflammation by recognizing double-strand RNA (dsRNA) released from UVB-irradiated cells. It has not been studied, however, whether apoptosis induction in UVB irradiation is attributed to TLR3 activation. Here, we report on the pro-apoptotic role of TLR3 in UVB-irradiated epidermal cells. Poly I:C, an analogue of dsRNA that activates TLR3, was used in combination with sub-lethal UVB (4.8 mJ/cm2) irradiation for investigating the effects of TLR3 activation on human immortalized keratinocyte HaCaT cells. Although sub-lethal dose of either Poly I:C or UVB alone did not induce cell death, UVB-Poly I:C co-treatment synergistically induced cell death by activation of caspase-3 and cleavages of ICAD and PARP, with apoptotic features when stained with Annexin V/PI or Hoechst 33342. Treatment with pan-caspase inhibitor, Z-VAD, attenuated UVB-Poly I:C-induced cell death. Silencing TLR3 by siRNA rescued HaCaT cells from UVB-Poly I:C-induced apoptosis. NF-κB, a major downstream component of TLR3 pathway, that usually negatively regulates the classical TLR3 apoptotic pathway, was analyzed by western blotting and immunofluorescence confocal microscopy. The results indicate to our surprise that NF-κB is translocated to nucleus in the cells co-treated with UVB-Poly I:C. The nuclear translocation of NF-κB is attenuated by TLR3 silencing. Treatment with BAY, an inhibitor of NF-κB pathway, blocked UVB-Poly I:C-induced apoptosis. Therefore, we conclude that NF-κB pathway plays a cytotoxic role in UVB-Poly I:C-treated HaCaT cells, mediating TLR3-related apoptosis.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , NF-kappa B/metabolism , Poly I-C/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Epidermal Cells/drug effects , Epidermal Cells/radiation effects , Humans , Keratinocytes/drug effects , Keratinocytes/radiation effects , RNA, Double-Stranded/drug effects , RNA, Double-Stranded/radiation effects , Skin/drug effects , Skin/radiation effects , Ultraviolet Rays
17.
Mol Cell Biol ; 11(7): 3719-25, 1991 Jul.
Article in English | MEDLINE | ID: mdl-1646394

ABSTRACT

A double-stranded RNA (dsRNA)-specific modification activity from Xenopus oocytes and human cells dsRNA modifier) converts adenosine residues present in dsRNA to inosines. The function of the dsRNA modifier is unknown, although it has been suggested that it may be part of the cellular antiviral response. We investigated the relationship between the activity of the dsRNA modifier, viral infection, and the antiviral response in human cells induced by poly(rI)-poly(rC) [poly(I.C)] treatment. We found, unexpectedly, that treatment of HeLa cells with poly(I.C) or other dsRNA molecules resulted in the dramatic inhibition of the dsRNA modifier. Mixing experiments, reconstruction experiments, and pretreatment of extracts with RNases indicated that inhibition of the dsRNA modifier did not result from the continued presence of a soluble inhibitor such as dsRNA) in the in vitro modification reactions. Treatment of cells with cyclohexamide or dactinomycin simultaneously with the poly(I.C) demonstrated that in vivo inhibition of the dsRNA modifier did not require new transcription or translation. The dsRNA modification activity was also substantially inhibited in cells infected with poliovirus and was slightly inhibited in cells infected with adenovirus. The inhibition of the dsRNA modifier during the antiviral state is thus not consistent with an antiviral function, and instead suggests another cellular function for dsRNA modification.


Subject(s)
Adenoviruses, Human/physiology , Poliovirus/physiology , Poly I-C/pharmacology , RNA, Double-Stranded/physiology , Adenoviruses, Human/drug effects , Cycloheximide/pharmacology , Dactinomycin/pharmacology , HeLa Cells/drug effects , HeLa Cells/physiology , Humans , Interferon Type I/pharmacology , Kinetics , Poliovirus/drug effects , RNA, Double-Stranded/drug effects , Transcription, Genetic
18.
Sci Rep ; 6: 21595, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26879672

ABSTRACT

(-)-Epigallocatechin-3-gallate (EGCG), a major polyphenol component of green tea, has recently been identified as an inhibitor of hepatitis C virus (HCV) entry. Here, we examined whether EGCG can enhance hepatocyte-mediated intracellular innate immunity against HCV. HCV dsRNAs (Core, E1-P7, NS-3'NTR and NS5A) induced interferon-λ1 (IFN-λ1) expression in human hepatocytes. These HCV dsRNAs also induced the expression of Toll-like receptor 3 (TLR3), retinoic acid-inducible gene I (RIG-I) and several antiviral IFN-stimulated genes (ISGs) expression. Although EGCG treatment of hepatocytes alone had little effect on TLR3 and RIG-I signaling pathways, EGCG significantly enhanced HCV dsRNAs-induced the expression of IFN-λ1, TLR3, RIG-I and antiviral ISGs in hepatocytes. Furthermore, treatment of HCV-infected hepatocytes with EGCG and HCV dsRNAs inhibited viral replication. Given that EGCG has the ability to enhance HCV dsRNAs-induced intracellular antiviral innate immunity against HCV, suggesting the potential application of EGCG as a new anti-HCV agent for HCV therapy.


Subject(s)
Antiviral Agents/administration & dosage , Catechin/analogs & derivatives , Hepacivirus/metabolism , Hepatocytes/metabolism , Hepatocytes/virology , Immunity, Innate/drug effects , RNA, Viral/metabolism , Antiviral Agents/metabolism , Catechin/administration & dosage , Catechin/metabolism , Cell Line, Tumor , DEAD Box Protein 58 , Hepatocytes/drug effects , Humans , Interferons , Interleukins/metabolism , RNA, Double-Stranded/drug effects , RNA, Double-Stranded/metabolism , Receptors, Immunologic , Signal Transduction/drug effects , Toll-Like Receptor 3
19.
Cell Death Differ ; 9(9): 981-94, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181749

ABSTRACT

Interferons enhance the cellular antiviral response by inducing expression of protective proteins. Many of these proteins are activated by dsRNA, a typical by-product of viral infection. Here we show that type-I and type-II interferons can sensitize cells to dsRNA-induced cytotoxicity. In caspase-8- or FADD-deficient Jurkat cells dsRNA induces necrosis, instead of apoptosis. In L929sA cells dsRNA-induced necrosis involves high reactive oxygen species production. The antioxidant butylated hydroxyanisole protects cells from necrosis, but shifts the response to apoptosis. Treatment with the caspase inhibitor benzyloxycarbonyl-Val-Ala-DL-Asp(OMe)-fluoromethylketone or overexpression of Bcl-2 prevent this shift and promote necrosis. Our results suggest that a single stimulus can initiate different death-signaling pathways, leading to either necrotic or apoptotic cell death. Inhibition of key events in these signaling pathways, such as caspase activation, cytochrome c release or mitochondrial reactive oxygen species production, tips the balance between necrosis and apoptosis, leading to dominance of one of these death programs.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Interferons/pharmacology , Jurkat Cells/drug effects , Necrosis , RNA Virus Infections/drug therapy , RNA Viruses/drug effects , RNA, Double-Stranded/drug effects , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/genetics , Butylated Hydroxyanisole/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/deficiency , Caspases/genetics , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Fas-Associated Death Domain Protein , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Jurkat Cells/metabolism , Jurkat Cells/virology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Virus Infections/genetics , RNA Viruses/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
20.
Pharmacol Ther ; 78(2): 55-113, 1998 May.
Article in English | MEDLINE | ID: mdl-9623881

ABSTRACT

The 2-5A system is an RNA degradation pathway that can be induced by the interferons (IFNs). Treatment of cells with IFN activates genes encoding several double-stranded RNA (dsRNA)-dependent synthetases. These enzymes generate 5'-triphosphorylated, 2',5'-phosphodiester-linked oligoadenylates (2-5A) from ATP. The effects of 2-5A in cells are transient since 2-5A is unstable in cells due to the activities of phosphodiesterase and phosphatase. 2-5A activates the endoribonuclease 2-5A-dependent RNase L, causing degradation of single-stranded RNA with moderate specificity. The human 2-5A-dependent RNase is an 83.5 kDa polypeptide that has little, if any, RNase activity, unless 2-5A is present. 2-5A binding to RNase L switches the enzyme from its off-state to its on-state. At least three 2',5'-linked oligoadenylates and a single 5'-phosphoryl group are required for maximal activation of the RNase. Even though the constitutive presence of 2-5A-dependent RNase is observed in nearly all mammalian cell types, cellular amounts of 2-5A-dependent mRNA and activity can increase after IFN treatment. One well-established role of the 2-5A system is as a host defense against some types of viruses. Since virus infection of cells results in the production and secretion of IFNs, and since dsRNA is both a frequent product of virus infection and an activator of 2-5A synthesis, the replication of encephalomyocarditis virus, which produces dsRNA during its life cycle, is greatly suppressed in IFN-treated cells as a direct result of RNA decay by the activated 2-5A-dependent RNase. This review covers the organic chemistry, enzymology, and molecular biology of 2-5A and its associated enzymes. Additional possible biological roles of the 2-5A system, such as in cell growth and differentiation, human immunodeficiency virus replication, heat shock, atherosclerotic plaque, pathogenesis of Type I diabetes, and apoptosis, are presented.


Subject(s)
Adenine Nucleotides/chemistry , Adenine Nucleotides/metabolism , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Cell Physiological Phenomena/drug effects , Interferons/pharmacology , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/metabolism , RNA, Double-Stranded/drug effects , Viruses/drug effects , Humans
SELECTION OF CITATIONS
SEARCH DETAIL