Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 377
Filter
1.
Environ Mol Mutagen ; 63(7): 320-328, 2022 08.
Article in English | MEDLINE | ID: mdl-36181379

ABSTRACT

Molnupiravir (MOV) is used to treat COVID-19. In cells, MOV is converted to the ribonucleoside analog N4-hydroxycytidine (NHC) and incorporated into the SARS-CoV-2 RNA genome during its replication, resulting in RNA mutations. The widespread accumulation of such mutations inhibits SARS-CoV-2 propagation. Although safety assessments by many regulatory agencies across the world have concluded that the genotoxic risks associated with the clinical use of MOV are low, concerns remain that it could induce DNA mutations in patients, particularly because numerous in vitro studies have shown that NHC is a DNA mutagen. In this study, we used HiFi sequencing, a technique that can detect ultralow-frequency substitution mutations in whole genomes, to evaluate the mutagenic effects of MOV in E. coli and of MOV and NHC in mouse lymphoma L5178Y cells and human lymphoblastoid TK6 cells. In all models, exposure to these compounds increased genome-wide mutation frequencies in a dose-dependent manner, and these increases were mainly composed of A:T → G:C transitions. The NHC exposure concentrations used for mammalian cells were comparable to those observed in the plasma of humans who received clinical doses of MOV.


Subject(s)
COVID-19 , Mutagens , Humans , Mice , Animals , Mutagens/toxicity , Escherichia coli/genetics , RNA, Viral/pharmacology , SARS-CoV-2 , DNA , Mammals/genetics
2.
Expert Rev Pharmacoecon Outcomes Res ; 22(8): 1215-1220, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36154357

ABSTRACT

BACKGROUND: In Russia, before 2022, the list of vital and essential drugs for HIV-infected patients previously untreated with antiretroviral drugs included the fixed-dose combination rilpivirine/tenofovir disoproxil fumarate/emtricitabine (RPV/TDF/FTC) but not doravirine/tenofovir disoproxil fumarate/lamivudine (DOR/TDF/3TC). METHODS: An indirect comparison of the efficacy of DOR/TDF/3TC and RPV/TDF/FTC defined by virologic suppression (HIV-1 RNA of <50 copies/mL at week 48) was made. The per-patient drug costs over 1 year were compared in a cost-minimization analysis. A budget impact analysis considered the costs to the healthcare system of including DOR/TDF/3TC as a treatment option for eligible patients in Russia over a 3-year time horizon. RESULTS: The indirect treatment comparison of DOR/TDF/3TC and RPV/TDF/FTC in treatment-naïve patients with baseline HIV-1 RNA 100,000 copies/ml or less showed no statistically significant difference (RR 0.914, 95% CI 0.833-1.003). In the cost-minimization analysis, the per-patient cost of one year of treatment with RPV/TDF/FTC and DOR/TDF/3TC was, respectively, ₽320,975 and ₽151,192, for a saving of ₽169,783. In the budget impact analysis, the adoption of DOR/TDF/3TC into clinical practice is expected to reduce drug costs by ₽333 million (23.8%) in year 3. CONCLUSIONS: Fixed-dose combination DOR/TDF/3TC is equally effective and cost-saving compared to RPV/TDF/FTC from Russian vital and essential drugs list perspective.


Subject(s)
Anti-HIV Agents , Drugs, Essential , HIV Infections , HIV Seropositivity , HIV-1 , Adult , Humans , HIV-1/genetics , Lamivudine/adverse effects , Tenofovir/pharmacology , Tenofovir/therapeutic use , Cost-Benefit Analysis , Viral Load , HIV Infections/drug therapy , RNA, Viral/pharmacology , RNA, Viral/therapeutic use , Emtricitabine/pharmacology , Emtricitabine/therapeutic use , HIV Seropositivity/drug therapy , Anti-HIV Agents/adverse effects , Drug Combinations
3.
J Pharmacol Sci ; 149(3): 81-84, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35641031

ABSTRACT

Ciclesonide (Cic) is approved as an inhalant for asthma and was clinically tested as a candidate therapy for coronavirus disease 2019 (COVID-19). Its active metabolite Cic2 was recently reported to suppress genomic RNA replication of severe acute respiratory syndrome coronavirus 2. In this study, we designed and synthesized a set of ciclesonide-acetal (Cic-acetal) derivatives. Among designated compounds, some Cic-acetal derivatives with a linear alkyl chain exhibited strong viral copy-number reduction activities compared with Cic2. These compounds might serve as lead compounds for developing novel anti-COVID-19 agents.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Acetals/pharmacology , Antiviral Agents/pharmacology , Humans , Pregnenediones , RNA, Viral/genetics , RNA, Viral/pharmacology , SARS-CoV-2 , Virus Replication/genetics
4.
J Antibiot (Tokyo) ; 75(6): 321-332, 2022 06.
Article in English | MEDLINE | ID: mdl-35440771

ABSTRACT

Staphylococcus aureus is one of the most dangerous pathogens commonly associated with high levels of morbidity and mortality. Sortase A is considered as a promising molecular target for the development of antistaphylococcal agents. Using hybrid virtual screening approach and FRET analysis, we have identified five compounds able to decrease the activity of sortase A by more than 50% at the concentration of 200 µM. The most promising compound was 2-(2-amino-3-chloro-benzoylamino)-benzoic acid which was able to inhibit S. aureus sortase A at the IC50 value of 59.7 µM. This compound was selective toward sortase A compared to other four cysteine proteases - cathepsin L, cathepsin B, rhodesain, and the SARS-CoV2 main protease. Microscale thermophoresis experiments confirmed that this compound bound sortase A with KD value of 189 µM. Antibacterial and antibiofilm assays also confirmed high specificity of the hit compound against two standard and three wild-type, S. aureus hospital infection isolates. The effect of the compound on biofilms produced by two S. aureus ATCC strains was also observed suggesting that the compound reduced biofilm formation by changing the biofilm structure and thickness.


Subject(s)
COVID-19 , Staphylococcal Infections , Aminoacyltransferases , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Biofilms , Cysteine Endopeptidases , Humans , Microbial Sensitivity Tests , RNA, Viral/pharmacology , SARS-CoV-2 , Staphylococcus aureus
5.
Nihon Yakurigaku Zasshi ; 157(2): 115-118, 2022.
Article in Japanese | MEDLINE | ID: mdl-35228442

ABSTRACT

In the renin-angiotensin system (RAS), angiotensin II (AngII) converted by angiotensin converting enzyme (ACE) exerts a strong physiological activity via the AT1 receptor (AT1R). Thus, the ACE-AngII-AT1R axis positively regulates RAS. On the other hand, angiotensin converting enzyme 2 (ACE2) is known to negatively regulate RAS by degrading AngII into angiotensin 1-7 (Ang1-7). In the acute respiratory distress syndrome (ARDS), which is characterized by pulmonary hyperinflammation, the AngII-AT1R axis acts to exacerbate ARDS and the ACE2-AT2R axis acts protectively. More recently, ACE2 has been shown to be a receptor for SARS-CoV, the causative virus of severe acute respiratory syndrome (SARS), and SARS-CoV2, the causative virus of the 2019 coronavirus infection (COVID-19). Therefore, inhibition of the binding between ACE2 and virus spike protein is a drug discovery target for antiviral drugs against SARS-CoV and SARS-CoV2. In addition, when SARS and COVID-19 become severe, ARDS with cytokine storm is occured. We reported that soluble ACE2 protein and microbial-derived ACE2 like enzyme suppress pulmonary hyperinflammation due to SARS and COVID-19, respectively. In addition, it has been reported that the ACE2-soluble protein has an effect of suppressing the establishment of infection by inhibiting the binding between SARS-CoV2 and the cell membrane surface ACE2. Here, we describe the role of ACE2 in the pathophysiology of SARS/COVID-19 from the perspectives of inhibiting the progression to ARDS by suppressing pulmonary inflammation and suppressing the replication of the virus by inhibiting the binding of ACE2 to the spike protein.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19 Drug Treatment , Humans , Peptidyl-Dipeptidase A/metabolism , Peptidyl-Dipeptidase A/pharmacology , RNA, Viral/metabolism , RNA, Viral/pharmacology , Renin-Angiotensin System/physiology , SARS-CoV-2
6.
PLoS One ; 16(12): e0260706, 2021.
Article in English | MEDLINE | ID: mdl-34871316

ABSTRACT

Airway epithelial barrier dysfunction is increasingly recognized as a key feature of asthma and other lung diseases. Respiratory viruses are responsible for a large fraction of asthma exacerbations, and are particularly potent at disrupting epithelial barrier function through pattern recognition receptor engagement leading to tight junction dysfunction. Although different mechanisms of barrier dysfunction have been described, relatively little is known about whether barrier integrity can be promoted to limit disease. Here, we tested three classes of drugs commonly prescribed to treat asthma for their ability to promote barrier function using a cell culture model of virus-induced airway epithelial barrier disruption. Specifically, we studied the corticosteroid budesonide, the long acting beta-agonist formoterol, and the leukotriene receptor antagonist montelukast for their ability to promote barrier integrity of a monolayer of human bronchial epithelial cells (16HBE) before exposure to the viral mimetic double-stranded RNA. Of the three, only budesonide treatment limited transepithelial electrical resistance and small molecule permeability (4 kDa FITC-dextran flux). Next, we used a mouse model of acute dsRNA challenge that induces transient epithelial barrier disruption in vivo, and studied the effects budesonide when administered prophylactically or therapeutically. We found that budesonide similarly protected against dsRNA-induced airway barrier disruption in the lung, independently of its effects on airway inflammation. Taken together, these data suggest that an under-appreciated effect of inhaled budesonide is to maintain or promote airway epithelial barrier integrity during respiratory viral infections.


Subject(s)
Asthma/drug therapy , Bronchi/drug effects , Bronchodilator Agents/pharmacology , Budesonide/pharmacology , Cell Membrane Permeability/drug effects , Poly I-C/antagonists & inhibitors , Acetates/pharmacology , Administration, Inhalation , Animals , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Bronchi/metabolism , Bronchi/pathology , Cell Line , Cyclopropanes/pharmacology , Dextrans/metabolism , Electric Impedance , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Formoterol Fumarate/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Mimicry , Poly I-C/pharmacology , Quinolines/pharmacology , RNA, Double-Stranded/antagonists & inhibitors , RNA, Double-Stranded/pharmacology , RNA, Viral/antagonists & inhibitors , RNA, Viral/pharmacology , Sulfides/pharmacology , Tight Junctions/drug effects , Tight Junctions/metabolism
7.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34453520

ABSTRACT

The human testis can be infected by a large number of RNA and DNA viruses. While various RNA virus infections may induce orchitis and impair testicular functions, DNA virus infection rarely affects the testis. Mechanisms underlying the differential effects of RNA and DNA viral infections on the testis remain unclear. In the current study, we therefore examined the effects of viral RNA and DNA sensor signaling pathways on mouse Sertoli cells (SC) and Leydig cells (LC). The local injection of viral RNA analogue polyinosinic-polycytidylic acid [poly(I:C)] into the testis markedly disrupted spermatogenesis, whereas the injection of the herpes simplex virus (HSV) DNA analogue HSV60 did not affect spermatogenesis. Poly(I:C) dramatically induced the expression of the proinflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 6 in SC and LC through Toll-like receptor 3 and interferon ß promoter stimulator 1 signaling pathways, impairing the integrity of the blood-testis barrier and testosterone synthesis. Poly(I:C)-induced TNF-α production thus plays a critical role in the impairment of cell functions. In contrast, HSV60 predominantly induced the expression of type 1 interferons and antiviral proteins via the DNA sensor signaling pathway, which did not affect testicular cell functions. Accordingly, the Zika virus induced high levels of TNF-α in SC and LC and impaired their respective cellular functions, whereas Herpes simplex virus type 2 principally induced antiviral responses and did not impair such functions. These results provide insights into the mechanisms by which RNA viral infections impair testicular functions.


Subject(s)
DNA, Viral/metabolism , Leydig Cells/metabolism , RNA, Viral/metabolism , Receptors, Virus/metabolism , Sertoli Cells/metabolism , Animals , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Chlorocebus aethiops , DNA, Viral/pharmacology , Leydig Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleic Acids/metabolism , Poly I-C/metabolism , Poly I-C/pharmacology , RNA, Viral/pharmacology , Sertoli Cells/drug effects , Signal Transduction/immunology , Testis/drug effects , Testis/metabolism , Vero Cells
8.
Nat Plants ; 6(6): 620-624, 2020 06.
Article in English | MEDLINE | ID: mdl-32483329

ABSTRACT

An in planta gene editing approach was developed wherein Cas9 transgenic plants are infected with an RNA virus that expresses single guide RNAs (sgRNAs). The sgRNAs are augmented with sequences that promote cell-to-cell mobility. Mutant progeny are recovered in the next generation at frequencies ranging from 65 to 100%; up to 30% of progeny derived from plants infected with a virus expressing three sgRNAs have mutations in all three targeted loci.


Subject(s)
Gene Editing/methods , Nicotiana/genetics , Plants, Genetically Modified/genetics , RNA Viruses/genetics , RNA, Guide, Kinetoplastida/pharmacology , RNA, Viral/pharmacology , Agrobacterium tumefaciens
9.
Front Immunol ; 11: 8, 2020.
Article in English | MEDLINE | ID: mdl-32038656

ABSTRACT

Strong innate and adaptive immune responses are paramount in combating viral infections. Dendritic cells (DCs) detect viral infections via cytosolic RIG-I like receptors (RLRs) RIG-I and MDA5 leading to MAVS-induced immunity. The DEAD-box RNA helicase DDX3 senses abortive human immunodeficiency virus 1 (HIV-1) transcripts and induces MAVS-dependent type I interferon (IFN) responses, suggesting that abortive HIV-1 RNA transcripts induce antiviral immunity. Little is known about the induction of antiviral immunity by DDX3-ligand abortive HIV-1 RNA. Here we synthesized a 58 nucleotide-long capped RNA (HIV-1 Cap-RNA58) that mimics abortive HIV-1 RNA transcripts. HIV-1 Cap-RNA58 induced potent type I IFN responses in monocyte-derived DCs, monocytes, macrophages and primary CD1c+ DCs. Compared with RLR agonist poly-I:C, HIV-1 Cap-RNA58 induced comparable levels of type I IFN responses, identifying HIV-1 Cap-RNA58 as a potent trigger of antiviral immunity. In monocyte-derived DCs, HIV-1 Cap-RNA58 activated the transcription factors IRF3 and NF-κB. Moreover, HIV-1 Cap-RNA58 induced DC maturation and the expression of pro-inflammatory cytokines. HIV-1 Cap-RNA58-stimulated DCs induced proliferation of CD4+ and CD8+ T cells and differentiated naïve T helper (TH) cells toward a TH2 phenotype. Importantly, treatment of DCs with HIV-1 Cap-RNA58 resulted in an efficient antiviral innate immune response that reduced ongoing HIV-1 replication in DCs. Our data strongly suggest that HIV-1 Cap-RNA58 induces potent innate and adaptive immune responses, making it an interesting addition in vaccine design strategies.


Subject(s)
Adaptive Immunity , HIV Infections/immunology , HIV-1/genetics , Host Microbial Interactions/immunology , Immunity, Innate , RNA, Viral/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/virology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/virology , Monocytes/immunology , Monocytes/virology , NF-kappa B/metabolism , RNA, Viral/chemical synthesis , RNA, Viral/immunology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transcription, Genetic
10.
Gynecol Endocrinol ; 36(4): 346-350, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31595804

ABSTRACT

The aim of this study was to evaluate the effect of virus infection on estradiol (E2) production in human ovarian granulosa cells. Polyriboinosinic polyribocytidylic acid [Poly (I: C)], a synthetic analog of viral double stranded RNA that can be recognized by Toll like receptor 3 (TLR3), was used to imitate virus infection. Granulosa cells (GCs) obtained from patients undergoing in vitro fertilization and embryo transfer (IVF-ET) were cultured in vitro and treated with Poly (I: C), FSH, or both. Concentration of E2 was assayed by electrochemiluminescence. The mRNA and protein expression of TLR3 and aromatase were determined by real-time quantitative PCR (qPCR) and Western blot, respectively. The results showed that expression of TLR3 mRNA was significantly increased after Poly (I: C) stimulation. Poly (I: C) decreased E2 synthesis in FSH-treated GCs. Poly (I: C) inhibited the expression of aromatase in FSH-treated GCs. This study demonstrated that Poly (I: C) inhibits the synthesis of estradiol by granulosa cells under the stimulation of FSH, which might contribute to disturbance of follicular development and ovulation.


Subject(s)
Estradiol/metabolism , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/drug effects , Poly I-C/pharmacology , Adult , Cells, Cultured , Female , Granulosa Cells/metabolism , Granulosa Cells/virology , Humans , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Ovarian Follicle/virology , RNA, Viral/pharmacology , Virus Diseases/metabolism , Virus Diseases/pathology , Young Adult
11.
Front Immunol ; 10: 836, 2019.
Article in English | MEDLINE | ID: mdl-31057555

ABSTRACT

The activation of innate immunity by viral nucleic acids present in the cytoplasm plays an essential role in controlling viral infection in both immune and non-immune cells. The dsDNA and dsRNA viral mimics can stimulate the cytosolic nucleic acids sensors and activate the antiviral innate immunity. In this study, taking advantage of dsDNA and dsRNA viral mimics, we investigated the global transcriptome changes after the antiviral immunity activation in mouse embryonic fibroblasts. Results from our data identified a positive feedback up-regulation of sensors (e.g., Tlr2, Tlr3, Ddx58, cGAS), transducers (e.g., Traf2, Tbk1) and transcription factors (e.g., Irf7, Jun, Stat1, Stat2) in multiple pathways involved in detecting viral or microbial infections upon viral mimic stimulation. A group of genes involved in DNA damage response and DNA repair such as Parp9, Dtx3l, Rad52 were also up-regulated, implying the involvement of these genes in antiviral immunity. Molecular function analysis further showed that groups of helicase genes (e.g., Dhx58, Helz2), nuclease genes (e.g., Dnase1l3, Rsph10b), methyltransferase genes (e.g., histone methyltransferase Prdm9, Setdb2; RNA methyltransferase Mettl3, Mttl14), and protein ubiquitin-ligase genes (e.g., Trim genes and Rnf genes) were up-regulated upon antiviral immunity activation. In contrast, viral mimic stimulation down-regulated genes involved in a broad range of general biological processes (e.g., cell division, metabolism), cellular components (e.g., mitochondria and ribosome), and molecular functions (e.g., cell-cell adhesion, microtubule binding). In summary, our study provides valuable information about the global transcriptome changes upon antiviral immunity activation. The identification of novel groups of genes up-regulated upon antiviral immunity activation serves as useful resource for mining new antiviral sensors and effectors.


Subject(s)
DNA, Viral/immunology , Embryo, Mammalian/immunology , Fibroblasts/immunology , Immunity, Innate , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Transcriptome/immunology , Animals , DNA, Viral/pharmacology , Embryo, Mammalian/cytology , Mice , RNA, Double-Stranded/pharmacokinetics , RNA, Viral/pharmacology , Transcriptome/drug effects
12.
Elife ; 82019 02 08.
Article in English | MEDLINE | ID: mdl-30735121

ABSTRACT

MicroRNA-122 (miR-122) is the most abundant microRNA in hepatocytes and a central player in liver biology and disease. Herein, we report a previously unknown role for miR-122 in hepatocyte intrinsic innate immunity. Restoration of miR-122 levels in hepatoma cells markedly enhanced the activation of interferons (IFNs) in response to a variety of viral nucleic acids or simulations, especially in response to hepatitis C virus RNA and poly (I:C). Mechanistically, miR-122 downregulated the phosphorylation (Tyr705) of STAT3, thereby removing the negative regulation of STAT3 on IFN-signaling. STAT3 represses IFN expression by inhibiting interferon regulatory factor 1 (IRF1), whereas miR-122 targets MERTK, FGFR1 and IGF1R, three receptor tyrosine kinases (RTKs) that directly promote STAT3 phosphorylation. This work identifies a miR-122-RTKs/STAT3-IRF1-IFNs regulatory circuitry, which may play a pivotal role in regulating hepatocyte innate immunity. These findings renewed our knowledge of miR-122's function and have important implications for the treatment of hepatitis viruses.


Subject(s)
Hepatitis/genetics , Immunity, Innate/genetics , Interferon Regulatory Factor-1/genetics , MicroRNAs/genetics , STAT3 Transcription Factor/genetics , Cell Line, Tumor , Hepacivirus/genetics , Hepacivirus/pathogenicity , Hepatitis/pathology , Hepatitis/virology , Hepatocytes/metabolism , Hepatocytes/virology , Host-Pathogen Interactions/genetics , Humans , Interferons/genetics , Liver/metabolism , Liver/pathology , Liver/virology , RNA, Viral/pharmacology , Signal Transduction , Virus Replication/genetics
13.
Front Med ; 11(4): 502-508, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29170915

ABSTRACT

Although the efficacy of nucleos(t)ide analogue (NA) has been confirmed for treatment of chronic hepatitis B, long-term therapy has been recommended due to the high frequency of off-therapy viral DNA rebound and disease relapse. In this review, the RNA virion-like particles of hepatitis B virus (HBV) are integrated into the life cycle of HBV replication, and the potential significance of serum HBV RNA is systematically described. The production of HBV RNA virion-like particles should not be blocked by NA; in this regard, serum HBV RNA is found to be a suitable surrogate marker for the activity of intrahepatic covalently closed circular DNA (cccDNA), particularly among patients receiving NA therapy. Therefore, the concept of virological response is redefined as persistent loss of serum HBV DNA and HBV RNA. In contrast to hepatitis B surface antigen (HBsAg) that can originate from either the cccDNA or the integrated HBV DNA fragment, serum HBV RNA, with pregenomic RNA origination, can only be transcribed from cccDNA. Therefore, the loss of serum HBV RNA would likely be a promising predicator for safe drug discontinuation. The clinical status of consistent loss of serum HBV RNA accompanied with low serum HBsAg levels might be implicated as a "para-functional cure," a status nearly close to the functional cure of chronic hepatitis B, to distinguish the "functional cure" characterized as serum HBsAg loss with or without anti-HBs seroconversion.


Subject(s)
Hepatitis B virus , Hepatitis B, Chronic , Nucleosides/pharmacology , RNA, Viral/pharmacology , Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/therapy , Hepatitis B, Chronic/virology , Humans , Treatment Outcome
14.
Acta Biomater ; 64: 237-248, 2017 12.
Article in English | MEDLINE | ID: mdl-29030308

ABSTRACT

Intranasal mRNA vaccination provides immediate immune protection against pandemic diseases. Recent studies have shown that diverse forms of polyethyleneimine (PEI) have potent mucosal adjuvant activity, which could significantly facilitate the delivery of intranasal mRNA vaccines. Nevertheless, optimizing the chemical structure of PEI to maximize its adjuvanticity and decrease its toxicity remains a challenge. Here we show that the chemical structure of PEI strongly influences how well nanocomplexes of PEI and mRNA migrate to the lymph nodes and elicit immune responses. Conjugating cyclodextrin (CD) with PEI600 or PEI2k yielded CP (CD-PEI) polymers with different CD/PEI ratios. We analyzed the delivery efficacy of CP600, CP2k, and PEI25k as intranasal mRNA vaccine carriers by evaluating the lymph nodes migration and immune responses. Among these polymers, CP2k/mRNA showed significantly higher in vitro transfection efficiency, stronger abilities to migrate to lymph nodes and stimulate dendritic cells maturation in vivo, which further led to potent humoral and cellular immune responses, and showed lower local and systemic toxicity than PEI25k/mRNA. These results demonstrate the potential of CD-PEI2k/mRNA nanocomplex as a self-adjuvanting vaccine delivery vehicle that traffics to lymph nodes with high efficiency. STATEMENT OF SIGNIFICANCE: As we face outbreaks of pandemic diseases such as Zika virus, intranasal mRNA vaccination provides instant massive protection against highly variant viruses. Various polymer-based delivery systems have been successfully applied in intranasal vaccine delivery. However, the influence of molecular structure of the polymeric carriers on the lymph node trafficking and dendritic cell maturation is seldom studied for intranasal vaccination. Therefore, engineering polymer-based vaccine delivery system and elucidating the relationship between molecular structure and the intranasal delivery efficiency are essential for maximizing the immune responses. We hereby construct self-adjuvanting polymer-based intranasal mRNA vaccines to enhance lymph node trafficking and further improve immune responses.


Subject(s)
Drug Carriers , RNA, Messenger , RNA, Viral , Transfection/methods , Vaccines , Zika Virus Infection , Zika Virus , Administration, Intranasal , Animals , Dogs , Drug Carriers/chemistry , Drug Carriers/pharmacology , Female , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , RNA, Messenger/chemistry , RNA, Messenger/immunology , RNA, Messenger/pharmacology , RNA, Viral/chemistry , RNA, Viral/immunology , RNA, Viral/pharmacology , Vaccines/chemistry , Vaccines/immunology , Vaccines/pharmacology , Zika Virus Infection/immunology , Zika Virus Infection/prevention & control
15.
Viruses ; 9(5)2017 05 19.
Article in English | MEDLINE | ID: mdl-28534856

ABSTRACT

Cytomegalovirus (CMV) infection leads to notable morbidity and mortality in immunosuppressed patients. Current antiviral drugs are effective but seriously limited in their long-term use due to their relatively high toxicity. In the present study, we characterized the expression of murine CMV microRNAs (MCMV miRNAs) both in vitro and in vivo. Although 29 miRNAs were detectable during in vitro infection, only 11 miRNAs (classified as Group 1) were detectable during in vivo infection, and as many as 18 viral miRNAs (classified as Group 2) were less detectable (<50% of animals) in both the liver and lungs. In addition, viral miRNA profiles in the blood revealed unstable and reduced expression. We next explored the in vitro effects of viral miRNAs on MCMV replication. The inhibition of Group 1 viral miRNAs had little effect on virus production, but transfected cells overexpressing miR-m01-3-5p, miR-M23-1-5p, miR-M55-1, and miR-m107-1-5p in Group 2 showed statistically lower viral loads than those transfected with control miRNA (29%, 29%, 39%, and 43%, respectively, versus control). Finally, we performed hydrodynamic injection of viral miRNA agomirs and observed lower levels of MCMV recurrence in the livers of animals overexpressing the miR-m01-3-5p or mcmv-miR-M23-1-5p agomirs compared with those of animals transfected with control agomir, confirming the antiviral effects of viral miRNA manipulation in vivo. Therefore, the manipulation of viral miRNA expression shows great therapeutic potential and represents a novel antiviral strategy for the miRNA-based treatment of cytomegalovirus infection.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , MicroRNAs/pharmacology , Muromegalovirus/genetics , RNA, Viral/pharmacology , Animals , Antigens, Viral , Cell Culture Techniques , Cytomegalovirus Infections/virology , Disease Models, Animal , Fibroblasts/virology , Gene Expression Regulation, Viral , Immunohistochemistry , Kinetics , Liver/pathology , Liver/virology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/isolation & purification , MicroRNAs/therapeutic use , Mouse Embryonic Stem Cells , Muromegalovirus/drug effects , RNA, Viral/genetics , RNA, Viral/isolation & purification , RNA, Viral/therapeutic use , Transfection , Viral Load , Virus Replication/drug effects
16.
Stem Cells ; 35(5): 1197-1207, 2017 05.
Article in English | MEDLINE | ID: mdl-28276156

ABSTRACT

We have revealed a critical role for innate immune signaling in nuclear reprogramming to pluripotency, and in the nuclear reprogramming required for somatic cell transdifferentiation. Activation of innate immune signaling causes global changes in the expression and activity of epigenetic modifiers to promote epigenetic plasticity. In our previous articles, we focused on the role of toll-like receptor 3 (TLR3) in this signaling pathway. Here, we define the role of another innate immunity pathway known to participate in response to viral RNA, the retinoic acid-inducible gene 1 receptor (RIG-1)-like receptor (RLR) pathway. This pathway is represented by the sensors of viral RNA, RIG-1, LGP2, and melanoma differentiation-associated protein 5 (MDA5). We first found that TLR3 deficiency only causes a partial inhibition of nuclear reprogramming to pluripotency in mouse tail-tip fibroblasts, which motivated us to determine the contribution of RLR. We found that knockdown of interferon beta promoter stimulator 1, the common adaptor protein for the RLR family, substantially reduced nuclear reprogramming induced by retroviral or by modified messenger RNA expression of Oct 4, Sox2, KLF4, and c-MYC (OSKM). Importantly, a double knockdown of both RLR and TLR3 pathway led to a further decrease in induced pluripotent stem cell (iPSC) colonies suggesting an additive effect of both these pathways on nuclear reprogramming. Furthermore, in murine embryonic fibroblasts expressing a doxycycline (dox)-inducible cassette of the genes encoding OSKM, an RLR agonist increased the yield of iPSCs. Similarly, the RLR agonist enhanced nuclear reprogramming by cell permeant peptides of the Yamanaka factors. Finally, in the dox-inducible system, RLR activation promotes activating histone marks in the promoter region of pluripotency genes. To conclude, innate immune signaling mediated by RLR plays a critical role in nuclear reprogramming. Manipulation of innate immune signaling may facilitate nuclear reprogramming to achieve pluripotency. Stem Cells 2017;35:1197-1207.


Subject(s)
Cellular Reprogramming/genetics , DEAD Box Protein 58/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Cellular Reprogramming/drug effects , Epigenesis, Genetic/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Ligands , Mice, Knockout , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , RNA, Viral/pharmacology , Tail , Toll-Like Receptor 3/metabolism
17.
Nat Plants ; 3: 16207, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28067898

ABSTRACT

Topical application of pathogen-specific double-stranded RNA (dsRNA) for virus resistance in plants represents an attractive alternative to transgenic RNA interference (RNAi). However, the instability of naked dsRNA sprayed on plants has been a major challenge towards its practical application. We demonstrate that dsRNA can be loaded on designer, non-toxic, degradable, layered double hydroxide (LDH) clay nanosheets. Once loaded on LDH, the dsRNA does not wash off, shows sustained release and can be detected on sprayed leaves even 30 days after application. We provide evidence for the degradation of LDH, dsRNA uptake in plant cells and silencing of homologous RNA on topical application. Significantly, a single spray of dsRNA loaded on LDH (BioClay) afforded virus protection for at least 20 days when challenged on sprayed and newly emerged unsprayed leaves. This innovation translates nanotechnology developed for delivery of RNAi for human therapeutics to use in crop protection as an environmentally sustainable and easy to adopt topical spray.


Subject(s)
Aluminum Silicates/pharmacology , Nanostructures/chemistry , Plant Diseases/prevention & control , Plant Viruses/drug effects , RNA Interference , RNA, Double-Stranded/pharmacology , RNA, Viral/pharmacology , Arabidopsis/physiology , Clay , Plant Diseases/virology , Plant Viruses/genetics , Nicotiana/physiology , Vigna/physiology
18.
Am J Respir Cell Mol Biol ; 56(4): 506-520, 2017 04.
Article in English | MEDLINE | ID: mdl-27911568

ABSTRACT

Airway remodeling is resultant of a complex multicellular response associated with a progressive decline of pulmonary function in patients with chronic airway disease. Here, repeated infections with respiratory viruses are linked with airway remodeling through largely unknown mechanisms. Although acute activation of the Toll-like receptor (TLR) 3 pathway by extracellular polyinosinic:polycytidylic acid (poly[I:C]) induces innate signaling through the NF-κB transcription factor in normal human small airway epithelial cells, prolonged (repetitive or tonic) poly(I:C) stimulation produces chronic stress fiber formation, mesenchymal transition, and activation of a fibrotic program. Chronic poly(I:C) stimulation enhanced the expression of core mesenchymal regulators Snail family zinc finger 1, zinc finger E-box binding homeobox, mesenchymal intermediate filaments (vimentin), and extracellular matrix proteins (fibronectin-1), and collagen 1A. This mesenchymal transition was prevented by silencing expression of NF-κB/RelA or administration of a small-molecule inhibitor of the IκB kinase, BMS345541. Acute poly(I:C) exposure in vivo induced profound neutrophilic airway inflammation. When administered repetitively, poly(I:C) resulted in enhanced fibrosis observed by lung micro-computed tomography, second harmonic generation microscopy of optically cleared lung tissue, and by immunohistochemistry. Epithelial flattening, expansion of the epithelial mesenchymal trophic unit, and enhanced Snail family zinc finger 1 and fibronectin 1 expression in airway epithelium were also observed. Repetitive poly(I:C)-induced airway remodeling, fibrosis, and epithelial-mesenchymal transition was inhibited by BMS345541 administration. Based on this novel model of viral inflammation-induced remodeling, we conclude that NF-κB is a major controller of epithelial-mesenchymal transition and pulmonary fibrosis, a finding that has potentially important relevance to airway remodeling produced by repetitive viral infections.


Subject(s)
Airway Remodeling , Epithelial-Mesenchymal Transition , Mesoderm/pathology , NF-kappa B/metabolism , Pneumonia/pathology , Pneumonia/physiopathology , Pulmonary Fibrosis/physiopathology , RNA, Viral/pharmacology , Airway Remodeling/drug effects , Animals , Bronchoalveolar Lavage Fluid , Chronic Disease , Collagen/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung/pathology , Mesoderm/drug effects , Mice, Inbred C57BL , Neutrophils/pathology , Pneumonia/complications , Pneumonia/diagnostic imaging , Poly I-C/pharmacology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Toll-Like Receptor 3/metabolism , X-Ray Microtomography
19.
Mol Ther ; 24(1): 135-45, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26548591

ABSTRACT

Inactivated Sendai virus (hemagglutinating virus of Japan; HVJ) envelope (HVJ-E) induces anticancer immunity and cancer cell-selective apoptosis through the recognition of viral RNA genome fragments by retinoic acid-inducible gene-I (RIG-I). Here, we discovered that the "copy-back" type of defective-interfering (DI) particles that exist in the Cantell strain of HVJ induced the human PC3 prostate cancer cell death more effectively than the Sendai/52 strain or Cantell strain, which contain fewer DI particles. DI particle genomic RNA (~550 bases) activated proapoptotic genes such as Noxa and/or TNF-related apoptosis-inducing ligand (TRAIL) in human prostate cancer cells to induce cancer cell-selective apoptosis. DI particle-derived RNA was synthesized by in vitro transcription (in vitro transcribed (IVT)-B2). IVT-B2 RNA, which has a double-stranded region in its secondary structure, promoted a stronger anticancer effect than IVT-HN RNA, which does not have a double-stranded region in its secondary structure. The intratumoral transfection of IVT-B2 significantly reduced the volume of a human prostate tumor and induced tumor cell apoptosis in the xenograft mouse model. Moreover, the involvement of natural killer (NK) cells in IVT-B2-RNA-induced anticancer effects was also suggested. These findings provide a novel nucleic acid medicine for the treatment of cancer.


Subject(s)
Defective Viruses/genetics , Prostatic Neoplasms/therapy , RNA, Double-Stranded/administration & dosage , RNA, Viral/administration & dosage , Sendai virus/genetics , Animals , Apoptosis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mitogen-Activated Protein Kinases , Oncolytic Virotherapy , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/pharmacology , RNA, Viral/chemistry , RNA, Viral/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays
20.
Eur J Pharm Biopharm ; 96: 437-41, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26423908

ABSTRACT

Due to the increased prevalence of resistant bacterial isolates which are no longer susceptible to antibiotic treatment, recent emphasis has been placed on finding alternative modes of treatment of wound infections. Bacteriophage have long been investigated for their antimicrobial properties, yet the utilization of phage therapy for the treatment of wound infections relies on a suitable delivery system. Poly(N-isopropylacrylamide) (PNIPAM) is a thermally responsive polymer which undergoes a temperature dependent phase transition at a critical solution temperature. Bacteriophage K has been successfully formulated with PNIPAM nanospheres copolymerized with allylamine (PNIPAM-co-ALA). By utilizing a temperature responsive polymer it has been possible to engineer the nanospheres to collapse at an elevated temperature associated with a bacterial skin infection. The nanogels were reacted with surface deposited maleic anhydride in order to anchor the nanogels to non-woven fabric. Bacteriophage incorporated PNIPAM-co-ALA nanospheres demonstrated successful bacterial lysis of a clinically relevant bacterial isolate - Staphylococcus aureus ST228 at 37°C, whilst bacterial growth was unaffected at 25°C, thus providing a thermally triggered release of bacteriophage.


Subject(s)
Acrylic Resins/chemistry , Anti-Bacterial Agents/chemistry , DNA, Viral/chemistry , Nanospheres/chemistry , RNA, Viral/chemistry , Staphylococcus aureus/drug effects , Adhesiveness , Administration, Cutaneous , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Bacteriolysis/drug effects , Bacteriophages/physiology , DNA, Viral/administration & dosage , DNA, Viral/pharmacology , Drug Carriers , Drug Compounding , Drug Liberation , Drug Resistance, Bacterial , Hot Temperature , Lysogeny , Maleic Anhydrides/chemistry , Nanospheres/ultrastructure , Phase Transition , RNA, Viral/administration & dosage , RNA, Viral/pharmacology , Staphylococcus aureus/growth & development , Staphylococcus aureus/virology , Surface Properties , Viral Tropism
SELECTION OF CITATIONS
SEARCH DETAIL
...