Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Arq. ciências saúde UNIPAR ; 26(3): 1376-1397, set-dez. 2022.
Article in English | LILACS | ID: biblio-1414509

ABSTRACT

In December 2019, a new coronavirus originating from the city of Wuhan in China started an epidemic that brought many countries into chaos and despair. SARS-CoV-2, as identified, gave rise to the severe acute respiratory syndrome called COVID-19. Its transmission happens through droplets of saliva, hand or contaminated surfaces. Since its discovery, COVID-19 has led many to death, therefore, researchers from around the world have joined efforts to develop strategies to contain the virus. In this race, drugs such as Chloroquine and Hydroxychloroquine have become possible options for showing an antiviral effect, however, studies contest their efficiency, generating uncertainties. Therefore, other alternatives have been investigated in this context, and the study of medicinal plants has been the target of research for the treatment of COVID-19 in search of bioactive natural products that can exert an antiviral action. The study aimed to analyze the published literature on COVID-19 (SARS-CoV-2) and its relationship with medicinal plants. Bibliographical survey. So far, no specific treatment against the disease has been found, only supportive, with drugs that aim to improve the individual's immune system and ensure that the virus does not replicate, for example, there are options such as chloroquine, hydroxychloroquine, remdesivir and convalescent plasma. On the other hand, studies have revealed that medicinal plants such as garlic, among others, showed efficiency in modulating proteins with a view to preventing viral replication and improving immunity against COVID-19. So far, there are no drugs that are completely safe and have been shown to have activity against the new coronavirus (SARS-CoV-2). However, medicinal plants can contribute to the development of specific therapies against SARS-CoV-2 in a safe and effective way.


Em dezembro de 2019, um novo coronavírus originário da cidade de Wuhan, na China, iniciou uma epidemia que levou muitos países ao caos e ao desespero. O SARS-CoV-2, conforme identificado, deu origem à síndrome respiratória aguda grave chamada COVID-19. Sua transmissão acontece através de gotículas de saliva, mãos ou superfícies contaminadas. Desde sua descoberta, o COVID-19 levou muitos à morte, por isso, pesquisadores de todo o mundo uniram esforços para desenvolver estratégias para conter o vírus. Nesta corrida, medicamentos como Cloroquina e Hidroxicloroquina tornaram-se opções possíveis por apresentarem efeito antiviral, porém, estudos contestam sua eficiência, gerando incertezas. Portanto, outras alternativas têm sido investigadas nesse contexto, e o estudo de plantas medicinais tem sido alvo de pesquisas para o tratamento da COVID- 19 em busca de produtos naturais bioativos que possam exercer ação antiviral. O estudo teve como objetivo analisar a literatura publicada sobre COVID-19 (SARS-CoV-2) e sua relação com plantas medicinais. Levantamento bibliográfico. Até o momento, não foi encontrado nenhum tratamento específico contra a doença, apenas de suporte, com medicamentos que visam melhorar o sistema imunológico do indivíduo e garantir que o vírus não se replique, por exemplo, há opções como cloroquina, hidroxicloroquina, remdesivir e convalescença plasma. Por outro lado, estudos revelaram que plantas medicinais como o alho, entre outras, mostraram eficiência na modulação de proteínas visando prevenir a replicação viral e melhorar a imunidade contra a COVID-19. Até o momento, não existem medicamentos completamente seguros e que tenham demonstrado atividade contra o novo coronavírus (SARS-CoV-2). No entanto, as plantas medicinais podem contribuir para o desenvolvimento de terapias específicas contra o SARS-CoV-2 de forma segura e eficaz.


En diciembre de 2019, un nuevo coronavirus originario de la ciudad de Wuhan, en China, inició una epidemia que sumió a muchos países en el caos y la desesperación. El SARS-CoV- 2, tal y como fue identificado, dio lugar al síndrome respiratorio agudo severo denominado COVID-19. Su transmisión se produce a través de gotitas de saliva, de las manos o de superficies contaminadas. Desde su descubrimiento, el COVID-19 ha llevado a muchos a la muerte, por lo que investigadores de todo el mundo han aunado esfuerzos para desarrollar estrategias de contención del virus. En esta carrera, fármacos como la Cloroquina y la Hidroxicloroquina se han convertido en posibles opciones por mostrar un efecto antiviral, sin embargo, los estudios refutan su eficacia, generando incertidumbres. Por lo tanto, otras alternativas han sido investigadas en este contexto, y el estudio de las plantas medicinales ha sido el objetivo de la investigación para el tratamiento de COVID-19 en busca de productos naturales bioactivos que puedan ejercer una acción antiviral. El estudio tuvo como objetivo analizar la literatura publicada sobre el COVID-19 (SARS-CoV-2) y su relación con las plantas medicinales. Estudio bibliográfico. Hasta el momento, no se ha encontrado un tratamiento específico contra la enfermedad, sólo de soporte, con fármacos que buscan mejorar el sistema inmunológico del individuo y asegurar que el virus no se replique, por ejemplo, existen opciones como la cloroquina, hidroxicloroquina, remdesivir y plasma convaleciente. Por otro lado, estudios han revelado que plantas medicinales como el ajo, entre otras, mostraron eficacia en la modulación de proteínas con vistas a impedir la replicación viral y mejorar la inmunidad contra el COVID-19. Hasta el momento, no existen medicamentos que sean completamente seguros y que hayan demostrado tener actividad contra el nuevo coronavirus (SARS-CoV-2). Sin embargo, las plantas medicinales pueden contribuir al desarrollo de terapias específicas contra el SARS-CoV-2 de forma segura y eficaz.


Subject(s)
Plants, Medicinal/immunology , SARS-CoV-2/drug effects , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/therapeutic use , Viral Vaccines/antagonists & inhibitors , Chloroquine/therapeutic use , Severe Acute Respiratory Syndrome/virology , Severe acute respiratory syndrome-related coronavirus , Pandemics/prevention & control , Garlic/immunology , COVID-19/epidemiology , Hydroxychloroquine/therapeutic use
2.
Int J Mol Sci ; 22(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33806254

ABSTRACT

The global COVID-19 pandemic caused by SARS-CoV-2 has resulted in over 2.2 million deaths. Disease outcomes range from asymptomatic to severe with, so far, minimal genotypic change to the virus so understanding the host response is paramount. Transcriptomics has become incredibly important in understanding host-pathogen interactions; however, post-transcriptional regulation plays an important role in infection and immunity through translation and mRNA stability, allowing tight control over potent host responses by both the host and the invading virus. Here, we apply ribosome profiling to assess post-transcriptional regulation of host genes during SARS-CoV-2 infection of a human lung epithelial cell line (Calu-3). We have identified numerous transcription factors (JUN, ZBTB20, ATF3, HIVEP2 and EGR1) as well as select antiviral cytokine genes, namely IFNB1, IFNL1,2 and 3, IL-6 and CCL5, that are restricted at the post-transcriptional level by SARS-CoV-2 infection and discuss the impact this would have on the host response to infection. This early phase restriction of antiviral transcripts in the lungs may allow high viral load and consequent immune dysregulation typically seen in SARS-CoV-2 infection.


Subject(s)
Cytokines/genetics , RNA Processing, Post-Transcriptional , Ribosomes/metabolism , Ribosomes/virology , SARS-CoV-2/immunology , Transcription Factors/genetics , Animals , Antiviral Agents/antagonists & inhibitors , Cell Line, Tumor , Chlorocebus aethiops , Computational Biology , Cytokines/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Profiling , Host Microbial Interactions , Humans , Immunity, Innate/genetics , Lung/immunology , Lung/virology , RNA, Messenger/metabolism , RNA-Seq , Ribosomes/genetics , SARS-CoV-2/metabolism , Transcription Factors/metabolism , Transcriptome , Vero Cells
3.
Biochem Pharmacol ; 186: 114436, 2021 04.
Article in English | MEDLINE | ID: mdl-33539815

ABSTRACT

Thrombocytopenia is common among patients with viral hepatitis, limiting the use of antiviral therapy. Eltrombopag (EP) is a thrombopoietin receptor (TPO-R) agonist that has been approved for treatment of immune thrombocytopenia patients with hepatitis virus infection. Interferon-α (IFN-α) plays a crucial role in the antiviral response, and is recommended as the first-line agent for chronic hepatitis B patients. Here, we investigated whether EP inhibits the production of IFN-stimulated genes (ISGs) induced by IFN-α through the TPO-R-independent pathway by mediating reactive oxygen species production by iron chelation. Our results assessed the inhibitory effect of EP on IFN-α signaling, which contributes to the downregulation of ISGs produced by monocytes and sheds light on the underlying mechanisms using iron chelation to treat patients with hepatitis-related immunological thrombocytopenia.


Subject(s)
Antiviral Agents/metabolism , Benzoates/pharmacology , Hydrazines/pharmacology , Interferon-alpha/metabolism , Iron/metabolism , Leukocytes, Mononuclear/metabolism , Pyrazoles/pharmacology , Adult , Animals , Antiviral Agents/antagonists & inhibitors , Benzoates/therapeutic use , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Female , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/metabolism , Humans , Hydrazines/therapeutic use , Interferon-alpha/antagonists & inhibitors , Leukocytes, Mononuclear/drug effects , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pyrazoles/therapeutic use , Receptors, Thrombopoietin/agonists , Receptors, Thrombopoietin/metabolism , THP-1 Cells/drug effects , THP-1 Cells/metabolism , Thrombocytopenia/drug therapy , Thrombocytopenia/metabolism
4.
Antiviral Res ; 187: 105018, 2021 03.
Article in English | MEDLINE | ID: mdl-33476709

ABSTRACT

MBX-2168 has a mechanism of action similar to that of acyclovir (ACV) and ganciclovir (GCV), but two unique steps differentiate this drug from ACV/GCV. First, MBX-2168 is, at least partially, phosphorylated by the endogenous cellular kinase TAOK3 to a monophosphate. The second involves the removal of a moiety at the 6-position of MBX-2168-MP by adenosine deaminase like protein-1 (ADAL-1). It has been previously demonstrated that co-incubation with pentostatin (dCF), an ADAL-1 inhibitor, antagonizes the anti-viral activity of MBX-2168. We therefore hypothesize that inhibiting ADAL-1 results in a reduction of active compound produced in virus-infected cells. To test this, we examined the effect dCF has on the conversion of MBX-2168 to a triphosphate in HSV-1 and HCMV-infected cells. Our results demonstrate incubation of MBX-2168 alone or with dCF in HCMV-infected cells resulted in 53.1 ± 0.7 and 39.4 ± 1.5 pmol triphosphate/106 cells at 120 h, respectively. Incubation of MBX-2168 alone or with dCF in Vero cells resulted in 12.8 ± 0.1 and 6.7 ± 0.7 pmol triphosphate/106 cells at 24 h, respectively. HSV-1-infected Vero cells demonstrated no statistical difference in triphosphate accumulation at 24 h (13.1 ± 0.3 pmol triphosphate/106 cells). As expected, incubation with dCF resulted in the accumulation of MBX-2168-MP in both HFF (9.8 ± 0.9 pmol MBX-2168-MP/106 cells at 120 h) and Vero cells (4.7 ± 0.3 pmol MBX-2168-MP/106 cells at 24 h) while no detectable levels of monophosphate were observed in cultures not incubated with dCF. We conclude that dCF antagonizes the anti-viral effect of MBX-2168 by inhibiting the production of triphosphate, the active compound.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Antiviral Agents/pharmacology , Cyclopropanes/antagonists & inhibitors , Cytomegalovirus/drug effects , Guanine/analogs & derivatives , Herpesvirus 1, Human/drug effects , Pentostatin/pharmacology , Polyphosphates/metabolism , Acyclovir/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Cyclopropanes/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Fibroblasts/virology , Foreskin/cytology , Ganciclovir/pharmacology , Guanine/antagonists & inhibitors , Guanine/pharmacology , Herpes Simplex/drug therapy , Herpes Simplex/virology , Host Microbial Interactions , Humans , Loss of Function Mutation , Male , Phosphorylation , Vero Cells , Virus Replication/drug effects
5.
PLoS Pathog ; 17(1): e1009183, 2021 01.
Article in English | MEDLINE | ID: mdl-33444388

ABSTRACT

The antiviral protein kinase R (PKR) is an important host restriction factor, which poxviruses must overcome to productively infect host cells. To inhibit PKR, many poxviruses encode a pseudosubstrate mimic of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2), designated K3 in vaccinia virus. Although the interaction between PKR and eIF2α is highly conserved, some K3 orthologs from host-restricted poxviruses were previously shown to inhibit PKR in a species-specific manner. To better define this host range function, we compared the sensitivity of PKR from 17 mammals to inhibition by K3 orthologs from closely related orthopoxviruses, a genus with a generally broader host range. The K3 orthologs showed species-specific inhibition of PKR and exhibited three distinct inhibition profiles. In some cases, PKR from closely related species showed dramatic differences in their sensitivity to K3 orthologs. Vaccinia virus expressing the camelpox virus K3 ortholog replicated more than three orders of magnitude better in human and sheep cells than a virus expressing vaccinia virus K3, but both viruses replicated comparably well in cow cells. Strikingly, in site-directed mutagenesis experiments between the variola virus and camelpox virus K3 orthologs, we found that different amino acid combinations were necessary to mediate improved or diminished inhibition of PKR derived from different host species. Because there is likely a limited number of possible variations in PKR that affect K3-interactions but still maintain PKR/eIF2α interactions, it is possible that by chance PKR from some potential new hosts may be susceptible to K3-mediated inhibition from a virus it has never previously encountered. We conclude that neither the sensitivity of host proteins to virus inhibition nor the effectiveness of viral immune antagonists can be inferred from their phylogenetic relatedness but must be experimentally determined.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Host Specificity , Orthopoxvirus/classification , Orthopoxvirus/physiology , Poxviridae Infections/virology , Virus Replication , eIF-2 Kinase/antagonists & inhibitors , Amino Acid Sequence , Animals , Antiviral Agents/metabolism , HeLa Cells , Humans , Phosphorylation , Phylogeny , Poxviridae Infections/genetics , Poxviridae Infections/metabolism , Sequence Homology , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
6.
J Virol ; 95(6)2021 02 24.
Article in English | MEDLINE | ID: mdl-33361422

ABSTRACT

DNA damage-inducible transcript 3 (DDIT3) plays important roles in endoplasmic reticulum (ER) stress-induced apoptosis and autophagy, but its role in innate immunity is not clear. Here, we report that DDIT3 inhibits the antiviral immune response during bovine viral diarrhea virus (BVDV) infection by targeting mitochondrial antiviral signaling (MAVS) in Madin-Darby bovine kidney (MDBK) cells and in mice. BVDV infection induced high DDIT3 mRNA and protein expression. DDIT3 overexpression inhibited type I interferon (IFN-I) and IFN-stimulated gene production, thereby promoting BVDV replication, while DDIT3 knockdown promoted the antiviral innate immune response to suppress viral replication. DDIT3 promoted NF-κB-dependent ovarian tumor (OTU) deubiquitinase 1 (OTUD1) expression. Furthermore, OTUD1 induced upregulation of the E3 ubiquitin ligase Smurf1 by deubiquitinating Smurf1, and Smurf1 degraded MAVS in MDBK cells in a ubiquitination-dependent manner, ultimately inhibiting IFN-I production. Moreover, knocking out DDIT3 promoted the antiviral innate immune response to reduce BVDV replication and pathological changes in mice. These findings provide direct insights into the molecular mechanisms by which DDIT3 inhibits IFN-I production by regulating MAVS degradation.IMPORTANCE Extensive studies have demonstrated roles of DDIT3 in apoptosis and autophagy during viral infection. However, the role of DDIT3 in innate immunity remains largely unknown. Here, we show that DDIT3 is positively regulated in bovine viral diarrhea virus (BVDV)-infected Madin-Darby bovine kidney (MDBK) cells and could significantly enhance BVDV replication. Importantly, DDIT3 induced OTU deubiquitinase 1 (OTUD1) expression by activating the NF-κB signaling pathway, thus increasing intracellular Smurf1 protein levels to degrade MAVS and inhibit IFN-I production during BVDV infection. Together, these results indicate that DDIT3 plays critical roles in host innate immunity repression and viral infection facilitation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Diarrhea Virus 1, Bovine Viral/physiology , Immunity, Innate , Transcription Factor CHOP/metabolism , Ubiquitin-Specific Proteases/metabolism , Virus Replication , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/immunology , Cattle , Diarrhea Virus 1, Bovine Viral/pathogenicity , Gene Expression Regulation , Host-Pathogen Interactions , Interferon Type I/antagonists & inhibitors , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , NF-kappa B/metabolism , Signal Transduction , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination
7.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32938761

ABSTRACT

SARS-CoV-2, a novel coronavirus (CoV) that causes COVID-19, has recently emerged causing an ongoing outbreak of viral pneumonia around the world. While distinct from SARS-CoV, both group 2B CoVs share similar genome organization, origins to bat CoVs, and an arsenal of immune antagonists. In this report, we evaluate type I interferon (IFN-I) sensitivity of SARS-CoV-2 relative to the original SARS-CoV. Our results indicate that while SARS-CoV-2 maintains similar viral replication to SARS-CoV, the novel CoV is much more sensitive to IFN-I. In Vero E6 and in Calu3 cells, SARS-CoV-2 is substantially attenuated in the context of IFN-I pretreatment, whereas SARS-CoV is not. In line with these findings, SARS-CoV-2 fails to counteract phosphorylation of STAT1 and expression of ISG proteins, while SARS-CoV is able to suppress both. Comparing SARS-CoV-2 and influenza A virus in human airway epithelial cultures, we observe the absence of IFN-I stimulation by SARS-CoV-2 alone but detect the failure to counteract STAT1 phosphorylation upon IFN-I pretreatment, resulting in near ablation of SARS-CoV-2 infection. Next, we evaluated IFN-I treatment postinfection and found that SARS-CoV-2 was sensitive even after establishing infection. Finally, we examined homology between SARS-CoV and SARS-CoV-2 in viral proteins shown to be interferon antagonists. The absence of an equivalent open reading frame 3b (ORF3b) and genetic differences versus ORF6 suggest that the two key IFN-I antagonists may not maintain equivalent function in SARS-CoV-2. Together, the results identify key differences in susceptibility to IFN-I responses between SARS-CoV and SARS-CoV-2 that may help inform disease progression, treatment options, and animal model development.IMPORTANCE With the ongoing outbreak of COVID-19, differences between SARS-CoV-2 and the original SARS-CoV could be leveraged to inform disease progression and eventual treatment options. In addition, these findings could have key implications for animal model development as well as further research into how SARS-CoV-2 modulates the type I IFN response early during infection.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Interferon Type I/pharmacology , Interferon-alpha/pharmacology , Severe acute respiratory syndrome-related coronavirus/drug effects , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/metabolism , Betacoronavirus/immunology , Betacoronavirus/physiology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon-alpha/antagonists & inhibitors , Interferon-alpha/immunology , Interferon-alpha/metabolism , Phosphorylation , Recombinant Proteins/pharmacology , Severe acute respiratory syndrome-related coronavirus/immunology , Severe acute respiratory syndrome-related coronavirus/physiology , SARS-CoV-2 , STAT1 Transcription Factor/metabolism , Signal Transduction , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
8.
J Virol ; 94(9)2020 04 16.
Article in English | MEDLINE | ID: mdl-32102878

ABSTRACT

Semen is the primary transmission vehicle for various pathogenic viruses. Initial steps of transmission, including cell attachment and entry, likely occur in the presence of semen. However, the unstable nature of human seminal plasma and its toxic effects on cells in culture limit the ability to study in vitro virus infection and inhibition in this medium. We found that whole semen significantly reduces the potency of antibodies and microbicides that target glycans on the envelope glycoproteins (Envs) of HIV-1. The extraordinarily high concentration of the monosaccharide fructose in semen contributes significantly to the effect by competitively inhibiting the binding of ligands to α1,2-linked mannose residues on Env. Infection and inhibition in whole human seminal plasma are accurately mimicked by a stable synthetic simulant of seminal fluid that we formulated. Our findings indicate that, in addition to the protein content of biological secretions, their small-solute composition impacts the potency of antiviral microbicides and mucosal antibodies.IMPORTANCE Biological secretions allow viruses to spread between individuals. Each type of secretion has a unique composition of proteins, salts, and sugars, which can affect the infectivity potential of the virus and inhibition of this process. Here, we describe HIV-1 infection and inhibition in whole human seminal plasma and a synthetic simulant that we formulated. We discovered that the sugar fructose in semen decreases the activity of a broad and potent class of antiviral agents that target mannose sugars on the envelope protein of HIV-1. This effect of semen fructose likely reduces the efficacy of such inhibitors to prevent the sexual transmission of HIV-1. Our findings suggest that the preclinical evaluation of microbicides and vaccine-elicited antibodies will be improved by their in vitro assessment in synthetic formulations that simulate the effects of semen on HIV-1 infection and inhibition.


Subject(s)
Fructose/metabolism , Fructose/pharmacology , Semen/metabolism , Adult , Anti-Infective Agents/pharmacology , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/pharmacology , Cell Line, Tumor , Gene Products, env/metabolism , Genes, env/genetics , HEK293 Cells , HIV Infections/virology , HIV-1/immunology , Humans , Male , Mannose/metabolism , Polysaccharides/immunology , Polysaccharides/metabolism , Semen/virology , env Gene Products, Human Immunodeficiency Virus/metabolism
9.
J Dairy Sci ; 102(11): 9559-9569, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31495632

ABSTRACT

Antiviral neuraminidase inhibitors, such as oseltamivir, zanamivir, and peramivir, are widely used for treatment of influenza virus infection. We reported previously that oseltamivir inhibits the viral growth cycle, ameliorates symptoms, and reduces viral antigen quantities. Suppressed viral antigen production, however, induces a reduction of acquired antiviral humoral immunity, and increases the incidence of re-infection rate in the following year. To achieve effective treatment of influenza virus infection, it is necessary to overcome these adverse effects of antiviral neuraminidase inhibitors. Feeding of yogurt fermented with Lactobacillus delbrueckii ssp. bulgaricus (L. bulgaricus) OLL1073R-1 is reported to have immune-stimulatory effects on influenza virus infection in mice and humans. In the present study, we assessed the effect of feeding L. bulgaricus OLL1073R-1 yogurt cultures (YC) on local and systemic humoral immune responses, which were suppressed by oseltamivir treatment, in mice infected with influenza A virus. Yogurt culture (1.14 × 108 cfu/0.4 mL per mouse per day) or sterile water (vehicle) was administered by intragastric gavage for 35 d. At d 22, influenza A virus/Puerto Rico/8/34 (H1N1) (PR8; 0.5 pfu/15 µL per mouse) was instilled intranasally, followed immediately by oral administration of oseltamivir (50 µg/100 µL per mouse, twice daily) or 5% methylcellulose (100 µL/mouse) as a vehicle for 13 d. Titers of anti-PR8-specific IgG and IgA in serum and mucosal secretory IgA (S-IgA) and IgG in bronchoalveolar lavage fluid (BALF) were analyzed by ELISA at 14 d after infection. Oseltamivir significantly suppressed the induction of anti-PR8-specific IgG and IgA in serum and S-IgA and IgG in BALF after infection. Feeding YC mildly but significantly stimulated production of PR8-specific IgA in serum, S-IgA in BALF, and IgG in serum without changing the IgG2a:IgG1 ratio. We analyzed the neutralizing activities against PR8 in serum and BALF and found that oseltamivir also reduced protective immunity, and YC feeding abrogated this effect. The immune-stimulatory tendency of YC on anti-PR8-specific IgA and IgG titers in serum and BALF was also detected in mice re-infected with PR8, but the effect was insignificant, unlike the effect of YC in the initial infection.


Subject(s)
Antiviral Agents/therapeutic use , Immunity, Humoral/drug effects , Lactobacillus delbrueckii , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae Infections/immunology , Oseltamivir/therapeutic use , Probiotics/therapeutic use , Viral Proteins/antagonists & inhibitors , Animal Feed , Animals , Antiviral Agents/adverse effects , Antiviral Agents/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Female , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Lactobacillus delbrueckii/drug effects , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/diet therapy , Orthomyxoviridae Infections/drug therapy , Oseltamivir/adverse effects , Oseltamivir/antagonists & inhibitors , Yogurt
10.
Nat Microbiol ; 4(6): 933-940, 2019 06.
Article in English | MEDLINE | ID: mdl-30886358

ABSTRACT

Type 1 interferon suppresses viral replication by upregulating the expression of interferon-stimulated genes with diverse antiviral properties1. The replication of human immunodeficiency virus type 1 (HIV-1) is naturally inhibited by interferon, with the steps between viral entry and chromosomal integration of viral DNA being notably susceptible2-5. The interferon-stimulated gene myxovirus resistance 2 has been defined as an effective postentry inhibitor of HIV-1, but is only partially responsible for interferon's suppressive effect6-8. Using small interfering RNA-based library screening in interferon-α-treated cells, we sought to characterize further interferon-stimulated genes that target the pre-integration phases of HIV-1 infection, and identified human tripartite-containing motif 5α (TRIM5α) as a potent anti-HIV-1 restriction factor. Human TRIM5α, in contrast with many nonhuman orthologues, has not generally been ascribed substantial HIV-1 inhibitory function, a finding attributed to ineffective recognition of cytoplasmic viral capsids by TRIM5α2,9,10. Here, we demonstrate that interferon-α-mediated stimulation of the immunoproteasome, a proteasome isoform mainly present in immune cells and distinguished from the constitutive proteasome by virtue of its different catalytic ß-subunits, as well as the proteasome activator 28 regulatory complex11-13, and the associated accelerated turnover of TRIM5α underpin the reprogramming of human TRIM5α for effective capsid-dependent inhibition of HIV-1 DNA synthesis and infection. These observations identify a mechanism for regulating human TRIM5α antiviral function in human cells and rationalize how TRIM5α participates in the immune control of HIV-1 infection.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Carrier Proteins/metabolism , HIV Infections/immunology , HIV-1/drug effects , Proteasome Endopeptidase Complex/metabolism , Antiviral Restriction Factors , Capsid/drug effects , Capsid Proteins/drug effects , Carrier Proteins/genetics , Cell Line , Gene Silencing , HEK293 Cells , Humans , Interferon-alpha/immunology , Interferon-alpha/metabolism , Myxovirus Resistance Proteins/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Virus Replication
11.
Eur J Pharm Biopharm ; 136: 147-155, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30682491

ABSTRACT

Chitosan is object of pharmaceutical research as a candidate permeability enhancer. However, chitosan was recently shown to reduce the oral bioavailability of acyclovir in humans. The effect of chitosan on two processes determining the oral bioavailability of acyclovir, bioaccessibility and intestinal absorption, was now investigated. Acyclovir's bioaccessibility was studied using the dynamic TNO gastro-Intestinal Model (TIM-1). Four epithelial models were used for permeability experiments: a Caco-2 cell model in absence and presence of mucus and both rat and porcine excised intestinal segments. Study concentrations of acyclovir (0.8 g/l) and chitosan (1.6 g/l and 4 g/l) were in line with those used in the aforementioned human study. No effect of chitosan was measured on the bioaccessibility of acyclovir in the TIM-1 system. The results obtained with the Caco-2 models were not in line with the in vivo data. The tissue segment models (rat and porcine intestine) showed a negative trend of acyclovir's permeation in presence of chitosan. The Ussing type chamber showed to be the most biopredictive, as it did point to an overall statistically significantly reduced absorption of acyclovir. This model thus seems most appropriate for pharmaceutical development purposes, in particular when interactions between excipients and drugs are to become addressed.


Subject(s)
Acyclovir/pharmacokinetics , Chitosan/pharmacokinetics , Intestinal Absorption/drug effects , Jejunum/metabolism , Acyclovir/administration & dosage , Acyclovir/antagonists & inhibitors , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/pharmacokinetics , Biocompatible Materials/administration & dosage , Biocompatible Materials/pharmacokinetics , Caco-2 Cells , Chitosan/administration & dosage , Drug Interactions/physiology , Humans , Intestinal Absorption/physiology , Jejunum/drug effects , Organ Culture Techniques , Permeability/drug effects , Rats , Swine
12.
J Med Virol ; 91(5): 803-812, 2019 05.
Article in English | MEDLINE | ID: mdl-30570770

ABSTRACT

Interleukin 6 (IL-6) is a pleiotropic cytokine with pivotal functions in the regulation of the biological responses of several target cells, including hepatocytes. Previous studies have shown that serum IL-6 levels are increased in hepatitis B patients. However, the role of IL-6 in modulating the anti-hepatitis B virus (HBV) activity of interferon-α (IFN-α) remains unclear. In this study, we found that both HBV and viral proteins could induce the expression of IL-6 in hepatocytes (LO2 and HepG2). Exogenous IL-6 had no effect on HBV replication, whereas knockdown of IL-6 expression by RNAi inhibited that. Interestingly, IFN-α markedly induced IL-6 expression in hepatocytes, especially in HBV replicating hepatocytes. In turn, IL-6 impaired the anti-HBV efficiency of IFN-α by decreases the expression of IFN-α downstream effectors by upregulation of suppressor of cytokine signaling-3 (SOCS3). Furthermore, we demonstrated that downregulation of SOCS3 improved IFN antiviral activity to some extent in HBV replicating hepatocytes. These data provided new insights for a better understanding of the mechanism of IFN-α resistance and may represent a novel therapeutic strategy to efficiently target HBV infection.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Hepatitis B virus/immunology , Host-Pathogen Interactions , Immune Evasion , Interferon-alpha/antagonists & inhibitors , Interleukin-6/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Cell Line , Hepatocytes/virology , Humans
13.
Microbiol Immunol ; 62(5): 341-347, 2018 May.
Article in English | MEDLINE | ID: mdl-29603339

ABSTRACT

Treatment of human embryonic lung fibroblast (HEL) cells with tricin (4', 5, 7-trihydroxy-3', 5'-dimethoxyflavone) following infection with human cytomegalovirus (HCMV) reportedly significantly suppresses HCMV replication. In the present work, the mechanisms for the anti-HCMV effects of tricin in HEL cells were examined. It was found that exposure of HEL cells to tricin inhibited HCMV replication, with concomitant decreases in amounts of transcripts of the CC chemokine RANTES (CCL5)-encoding gene and in expression of the CCL5 protein. It was also found that transcripts of HCMV immediate early 1 (IE1), and HCMV UL54 (encoding DNA polymerase) and replication of HCMV was significantly lower in CCL5 gene-knockdown cells. These results suggest that the anti-HCMV activity of tricin differs from that of ganciclovir and that CCL5 is one of the chemokines involved in HCMV replication. In addition, it is possible that chemokine CCL5 is one of the targets of tricin.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Chemokine CCL5/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus/growth & development , Flavonoids/antagonists & inhibitors , Gene Expression/drug effects , Virus Replication/drug effects , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , DNA Replication/drug effects , DNA-Directed DNA Polymerase , Fibroblasts/drug effects , Ganciclovir/antagonists & inhibitors , Gene Knockdown Techniques , Gene Silencing , Humans , Immediate-Early Proteins , RNA, Small Interfering , Transfection , Viral Proteins/genetics
14.
Trends Microbiol ; 26(5): 447-461, 2018 05.
Article in English | MEDLINE | ID: mdl-29395729

ABSTRACT

More than 100 pathogens, spanning multiple virus families, broadly termed 'arthropod-borne viruses (arboviruses)' have been associated with human and/or animal diseases. These viruses persist in nature through transmission cycles that involve alternating replication in susceptible vertebrate and invertebrate hosts. Collectively, these viruses are among the greatest burdens to global health, due to their widespread prevalence, and the severe morbidity and mortality they cause in human and animal hosts. Specific examples of mosquito-borne pathogens include Zika virus (ZIKV), West Nile virus (WNV), dengue virus serotypes 1-4 (DENV 1-4), Japanese encephalitis virus (JEV), yellow fever virus (YFV), chikungunya virus (CHIKV), and Rift Valley fever virus (RVFV). Interactions between arboviruses and the immune pathways of vertebrate hosts have been extensively reviewed. In this review we focus on the antiviral immune pathways present in mosquitoes. We also discuss mechanisms by which mosquito-borne viruses may antagonize antiviral pathways in disease vectors. Finally, we elaborate on the possibility that mosquito-borne viruses may be engaged in an evolutionary arms race with their invertebrate vector hosts, and the possible implications of this for understanding the transmission of mosquito-borne viruses.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Antiviral Agents/immunology , Arbovirus Infections/immunology , Arbovirus Infections/veterinary , Arboviruses/immunology , Culicidae/immunology , Mosquito Vectors/immunology , Adaptive Immunity , Animals , Arbovirus Infections/transmission , Chikungunya virus , Culicidae/virology , Dengue Virus , Encephalitis Virus, Japanese , Host-Pathogen Interactions/immunology , Humans , MicroRNAs/metabolism , Mosquito Vectors/virology , RNA Interference , RNA, Small Interfering/metabolism , Rift Valley fever virus , Virus Replication/immunology , Yellow fever virus , Zika Virus
15.
Virol J ; 14(1): 215, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29110727

ABSTRACT

BACKGROUND: Interferon (IFN) inhibits viruses by inducing several hundred cellular genes, aptly named 'interferon (IFN)-stimulated genes' (ISGs). The only two RNA viruses of the Pneumovirus genus of the Paramyxoviridae family, namely Respiratory Syncytial Virus (RSV) and Pneumonia Virus of Mice (PVM), each encode two nonstructural (NS) proteins that share no sequence similarity but yet suppress IFN. Since suppression of IFN underlies the ability of these viruses to replicate in the host cells, the mechanism of such suppression has become an important area of research. This Short Report is an important extension of our previous efforts in defining this mechanism. RESULTS: We show that, like their PVM counterparts, the RSV NS proteins also target multiple members of the ISG family. While significantly extending the substrate repertoire of the RSV NS proteins, these results, unexpectedly, also reveal that the target preferences of the NS proteins of the two viruses are entirely different. This is surprising since the two Pneumoviruses are phylogenetically close with similar genome organization and gene function, and the NS proteins of both also serve as suppressors of host IFN response. CONCLUSION: The finding that the NS proteins of the two highly similar viruses suppress entirely different members of the ISG family raises intriguing questions of pneumoviral NS evolution and mechanism of action.


Subject(s)
Murine pneumonia virus/physiology , Respiratory Syncytial Virus, Human/physiology , Viral Nonstructural Proteins/metabolism , Animals , Antiviral Agents/antagonists & inhibitors , Genetic Variation , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Interferons/antagonists & inhibitors , Mice , Substrate Specificity , Viral Nonstructural Proteins/genetics
16.
J Microbiol Biotechnol ; 27(11): 2070-2073, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-28910865

ABSTRACT

We have discovered a novel chemical compound, (E)-3-(furan-2-yl)-N-(4-sulfamoylphenyl) acrylamide, that suppresses the enzymatic activities of SARS coronavirus helicase. To determine the inhibitory effect, ATP hydrolysis and double-stranded DNA unwinding assays were performed in the presence of various concentrations of the compound. Through these assays, we obtained IC50 values of 2.09 ± 0.30 µM (ATP hydrolysis) and 13.2 ± 0.9 µM (DNA unwinding), respectively. Moreover, we found that the compound did not have any significant cytotoxicity when 40 µM of it was used. Our results showed that the compound might be useful to be developed as an inhibitor against SARS coronavirus.


Subject(s)
Antiviral Agents/antagonists & inhibitors , DNA Helicases/drug effects , Enzyme Inhibitors/pharmacology , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/enzymology , Adenosine Triphosphate , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cell Line/drug effects , Cell Survival/drug effects , DNA/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Humans , Hydrolysis , Inhibitory Concentration 50
17.
Article in English | MEDLINE | ID: mdl-28713784

ABSTRACT

Influenza A virus is a negative RNA stranded virus of the family Orthomyxoviridae, and represents a major public health threat, compounding existing disease conditions. Influenza A virus replicates rapidly within its host and the segmented nature of its genome facilitates re-assortment, whereby whole genes are exchanged between influenza virus subtypes during replication. Antiviral medications are important pharmacological tools in influenza virus prophylaxis and therapy. However, the use of currently available antiviral is impeded by sometimes high levels of resistance in circulating virus strains. Here, we identified novel anti-influenza compounds through screening of chemical compounds synthesized de novo on human lung epithelial cells. Computational and experimental screening of extensive and water soluble compounds identified novel influenza virus inhibitors that can reduce influenza virus infection without detectable toxic effects on host cells. Interestingly, the indicated active compounds inhibit viral replication most likely via interaction with cell receptors and disturb influenza virus entry into host cells. Collectively, screening of new synthesis chemical compounds on influenza A virus replication provides a novel and efficacious anti-influenza compounds that can inhibit viral replication via disturbing virus entry and indicates that these compounds are attractive candidates for evaluation as potential anti-influenza drugs.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Influenza A virus/drug effects , Orthomyxoviridae Infections/drug therapy , Virus Internalization/drug effects , A549 Cells , Animals , Antiviral Agents/chemical synthesis , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Drug Discovery , Drug Evaluation, Preclinical , Epithelial Cells/virology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/drug therapy , Lung/virology , Molecular Docking Simulation , Orthomyxoviridae Infections/virology , Virus Replication
18.
Antiviral Res ; 143: 218-229, 2017 07.
Article in English | MEDLINE | ID: mdl-28461069

ABSTRACT

The recent re-emergence of Zika virus (ZIKV)1, a member of the Flaviviridae family, has become a global emergency. Currently, there are no effective methods of preventing or treating ZIKV infection, which causes severe neuroimmunopathology and is particularly harmful to the developing fetuses of infected pregnant women. However, the pathology induced by ZIKV is unique among flaviviruses, and knowledge of the biology of other family members cannot easily be extrapolated to ZIKV. Thus, structure-function studies of ZIKV proteins are urgently needed to facilitate the development of effective preventative and therapeutic agents. Like other flaviviruses, ZIKV expresses an NS2B-NS3 protease, which consists of the NS2B cofactor and the NS3 protease domain and is essential for cleavage of the ZIKV polyprotein precursor and generation of fully functional viral proteins. Here, we report the enzymatic characterization of ZIKV protease, and we identify structural scaffolds for allosteric small-molecule inhibitors of this protease. Molecular modeling of the protease-inhibitor complexes suggests that these compounds bind to the druggable cavity in the NS2B-NS3 protease interface and affect productive interactions of the protease domain with its cofactor. The most potent compound demonstrated efficient inhibition of ZIKV propagation in vitro in human fetal neural progenitor cells and in vivo in SJL mice. The inhibitory scaffolds could be further developed into valuable research reagents and, ultimately, provide a roadmap for the selection of efficient inhibitors of ZIKV infection.


Subject(s)
Allosteric Site , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/chemistry , Zika Virus/enzymology , Amino Acid Sequence , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/chemistry , Base Sequence , Enzyme Activation , Female , Flavivirus/chemistry , Gene Expression , Humans , Inhibitory Concentration 50 , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , RNA Helicases/chemistry , RNA Helicases/drug effects , SOXB1 Transcription Factors/genetics , Sequence Alignment , Serine Endopeptidases/chemistry , Serine Endopeptidases/drug effects , Stem Cells , Viral Nonstructural Proteins/drug effects , Viral Proteins/chemistry , Viral Proteins/genetics , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus/growth & development , Zika Virus Infection/virology
19.
Microbiol Immunol ; 61(6): 232-238, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28463406

ABSTRACT

JC polyomavirus (JCPyV) is the causative agent of the demyelinating disease of the central nervous system known as progressive multifocal leukoencephalopathy (PML), which occurs in immunocompromised patients. Moreover, patients treated with natalizumab for multiple sclerosis or Crohn disease can develop PML, which is then termed natalizumab-related PML. Because few drugs are currently available for treating PML, many antiviral agents are being investigated. It has been demonstrated that the topoisomerase I inhibitors topotecan and ß-lapachone have inhibitory effects on JCPyV replication in IMR-32 cells. However, both of these drugs have marginal inhibitory effects on virus propagation in JC1 cells according to RT-PCR analysis. In the present study, the inhibitory effect of another topoisomerase I inhibitor, 7-ethy-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT11), was assessed by investigating viral replication, propagation, and viral protein 1 (VP1) production in cultured cells. JCPyV replication was assayed using real-time PCR combined with Dpn I treatment in IMR-32 cells transfected with JCPyV DNA. It was found that JCPyV replicates less in IMR-32 cells treated with CPT11 than in untreated cells. Moreover, CPT11 treatment of JCI cells persistently infected with JCPyV led to a dose-dependent reduction in JCPyV DNA and VP1 production. Additionally, the inhibitory effect of CPT11 was found to be stronger than those of topotecan and ß-lapachone. These findings suggest that CPT11 may be a potential anti-JCPyV agent that could be used to treat PML.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Camptothecin/antagonists & inhibitors , JC Virus/drug effects , Virus Replication/drug effects , Camptothecin/administration & dosage , Camptothecin/toxicity , Cell Line/drug effects , Cell Line/virology , Cell Proliferation/drug effects , DNA Replication/drug effects , DNA, Viral/genetics , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , JC Virus/genetics , Leukoencephalopathy, Progressive Multifocal/drug therapy , Naphthoquinones/antagonists & inhibitors , Real-Time Polymerase Chain Reaction/methods , Topoisomerase I Inhibitors/pharmacology , Topotecan/antagonists & inhibitors , Viral Proteins/drug effects
20.
Antiviral Res ; 143: 205-217, 2017 07.
Article in English | MEDLINE | ID: mdl-28450058

ABSTRACT

We previously reported low sensitivity of the hepatitis B virus (HBV) ribonuclease H (RNaseH) enzyme to inhibition by N-hydroxyisoquinolinedione (HID) compounds. Subsequently, our biochemical RNaseH assay was found to have a high false negative rate for predicting HBV replication inhibition, leading to underestimation of the number of HIDs that inhibit HBV replication. Here, 39 HID compounds and structurally related polyoxygenated heterocycles (POH), N-hydroxypyridinediones (HPD), and flutimides were screened for inhibition of HBV replication in vitro. Inhibiting the HBV RNaseH preferentially blocks synthesis of the positive-polarity DNA strand and causes accumulation of RNA:DNA heteroduplexes. Eleven HIDs and one HPD preferentially inhibited HBV positive-polarity DNA strand accumulation. EC50s ranged from 0.69 µM to 19 µM with therapeutic indices from 2.4 to 71. Neither the HIDs nor the HPD had an effect on the ability of the polymerase to elongate DNA strands in capsids. HBV RNaseH inhibition by the HIDs was confirmed with an improved RNaseH assay and by detecting accumulation RNA:DNA heteroduplexes in HBV capsids from cells treated with a representative HID. Therefore, the HID scaffold is more promising for anti-HBV drug discovery than we originally reported, and the HPD scaffold may hold potential for antiviral development. The preliminary structure-activity relationship will guide optimization of the HID/HPDs as HBV inhibitors.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Antiviral Agents/chemistry , Hepatitis B virus/drug effects , Virus Replication/drug effects , Animals , Antiviral Agents/administration & dosage , Capsid Proteins/genetics , Cell Line, Tumor , Chlorocebus aethiops , DNA Replication/drug effects , DNA, Viral/drug effects , Drug Discovery , Drug Evaluation, Preclinical , Hepatitis B/virology , Hepatitis B virus/enzymology , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , Microbial Sensitivity Tests , Piperazines/pharmacology , Ribonuclease H/drug effects , Structure-Activity Relationship , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...