Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Curr Med Chem ; 31(15): 2052-2072, 2024.
Article in English | MEDLINE | ID: mdl-37855348

ABSTRACT

Hepatitis C virus (HCV) is a globally prevalent and hazardous disorder that is responsible for inducing several persistent and potentially fatal liver diseases. Current treatment strategies offer limited efficacy, often accompanied by severe and debilitating adverse effects. Consequently, there is an urgent and compelling need to develop novel therapeutic interventions that can provide maximum efficacy in combating HCV while minimizing the burden of adverse effects on patients. One promising target against HCV is the NS3-4A serine protease, a complex composed of two HCV-encoded proteins. This non-covalent heterodimer is crucial in the viral life cycle and has become a primary focus for therapeutic interventions. Although peginterferon, combined with ribavirin, is commonly employed for HCV treatment, its efficacy is hampered by significant adverse effects that can profoundly impact patients' quality of life. In recent years, the development of direct-acting antiviral agents (DAAs) has emerged as a breakthrough in HCV therapy. These agents exhibit remarkable potency against the virus and have demonstrated fewer adverse effects when combined with other DAAs. However, it is important to note that there is a potential for developing resistance to DAAs due to alterations in the amino acid position of the NS3-4A protease. This emphasizes the need for ongoing research to identify strategies that can minimize the emergence of resistance and ensure long-term effectiveness. While the combination of DAAs holds promise for HCV treatment, it is crucial to consider the possibility of drug-drug interactions. These interactions may occur when different DAAs are used concurrently, potentially compromising their therapeutic efficacy. Therefore, carefully evaluating and monitoring potential drug interactions are vital to optimize treatment outcomes. In the pursuit of novel therapeutic interventions for HCV, the field of computational biology and bioinformatics has emerged as a valuable tool. These advanced technologies and methodologies enable the development and design of new drugs and therapeutic agents that exhibit maximum efficacy, reduced risk of resistance, and minimal adverse effects. By leveraging computational approaches, researchers can efficiently screen and optimize potential candidates, accelerating the discovery and development of highly effective treatments for HCV, treatments.


Subject(s)
Hepatitis C, Chronic , Hepatitis C , Humans , Hepacivirus , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Antiviral Agents/chemistry , Quality of Life , Hepatitis C, Chronic/drug therapy , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/pharmacology , Hepatitis C/drug therapy , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Protease Inhibitors/chemistry
2.
J Biol Inorg Chem ; 28(1): 101-115, 2023 02.
Article in English | MEDLINE | ID: mdl-36484824

ABSTRACT

Chikungunya virus (CHIKV) is the causative agent of chikungunya fever, a disease that can result in disability. Until now, there is no antiviral treatment against CHIKV, demonstrating that there is a need for development of new drugs. Studies have shown that thiosemicarbazones and their metal complexes possess biological activities, and their synthesis is simple, clean, versatile, and results in high yields. Here, we evaluated the mechanism of action (MOA) of a cobalt(III) thiosemicarbazone complex named [CoIII(L1)2]Cl based on its in vitro potent antiviral activity against CHIKV previously evaluated (80% of inhibition on replication). Furthermore, the complex has no toxicity in healthy cells, as confirmed by infecting BHK-21 cells with CHIKV-nanoluciferase in the presence of the compound, showing that [CoIII(L1)2]Cl inhibited CHIKV infection with the selective index of 3.26. [CoIII(L1)2]Cl presented a post-entry effect on viral replication, emphasized by the strong interaction of [CoIII(L1)2]Cl with CHIKV non-structural protein 4 (nsP4) in the microscale thermophoresis assay, suggesting a potential mode of action of this compound against CHIKV. Moreover, in silico analyses by molecular docking demonstrated potential interaction of [CoIII(L1)2]Cl with nsP4 through hydrogen bonds, hydrophobic and electrostatic interactions. The evaluation of ADME-Tox properties showed that [CoIII(L1)2]Cl presents appropriate lipophilicity, good human intestinal absorption, and has no toxicological effect as irritant, mutagenic, reproductive, and tumorigenic side effects.


Subject(s)
Chikungunya Fever , Chikungunya virus , Humans , Chikungunya Fever/drug therapy , Chikungunya Fever/metabolism , Chikungunya virus/metabolism , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/pharmacology , Viral Nonstructural Proteins/therapeutic use , Cobalt/pharmacology , Molecular Docking Simulation , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
3.
Article in English | MEDLINE | ID: mdl-36197422

ABSTRACT

The Hepatitis C virus (HCV) infection is a public health problem. The high level of HCV replication and its lack of post-transcriptional correction mechanisms results in the emergence of viral variants and the difficulty in determining polymorphisms and variants that contain the substitutions associated with resistance towards new antivirals. The main focus of this study was to map the NS5A and NS5B polymorphisms and resistance mutations to new antiviral drugs in HCV strains genotype 1 from patients with chronic hepatitis C infection. Serum samples were collected from patients who underwent routine viral load tests at the Instituto Adolfo Lutz, Sao Paulo city, Brazil. A total of 698 and 853 samples were used for the characterization of NS5A and NS5B regions, respectively, which comprise the HCV genotypes 1a and 1b. The prevalence of resistance mutations found in the NS5A region was 6.4%, with Y93H, L31M, Q30R, and Y93N as the main resistance-associated substitutions (RAS). No NS5B-associated RAS was observed for any of the analyzed drugs. These findings support that the RAS test should be offered to individuals with poor response to double combination regimens prior to treatment initiation, thereby assisting strain vigilance and selection of effective treatment or retreatment options using DAA regimens.


Subject(s)
Hepatitis C, Chronic , Hepatitis C , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Brazil , Drug Resistance, Viral/genetics , Genotype , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Humans , Mutation , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/pharmacology , Viral Nonstructural Proteins/therapeutic use
4.
J Appl Microbiol ; 133(5): 2826-2834, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35916643

ABSTRACT

AIMS: Pakistan has the second highest prevalence of HCV with genotype 3a (GT-3a) being the most frequently circulating genotype. Currently, resistance-associated substitutions (RASs) are a major challenge in HCV treatment with direct-acting antivirals (DAAs). Sofosbuvir (SOF) is an FDA-approved NS5B nucleotide inhibitor. The aim of this study was to identify these RASs in the NS5B gene in naive and treated Pakistani HCV 3a isolates against SOF. METHODS AND RESULTS: Blood samples were collected from anti-HCV-positive patients, followed by HCV RNA isolation and real-time PCR quantification. HCV-positive patients were processed for HCV RNA genotyping, patients with genotype 3a were processed for NS5B gene amplification and sequencing. GT-3a was the most prevalent genotype (62.2%). S282T was identified in 2 (8.7%) patients, C316Y/G/R in 3 (13%), V321A and L320P in 1 (4.3%) each in SOF/RBV-resistant patients. Variants of S282 were detected in 3 (13%) of SOF/RBV-treated patients. While INF/RBV-associated mutations were also analysed, D244N, A333R and A334E were identified in 2 (9.5%), 3 (14.2%) and 7 (33.3%) in treatment-naive and 15 (65.2%), 7 (30.4%) and 5 (21.7%) treated patients, respectively. Q309R was observed only in one treatment-experienced patients. Some substitutions were present at higher frequency in both groups like N307G, K304R, A272D and R345H, considered that they do not have any role in sofosbuvir resistance. CONCLUSION: It was concluded that sofosbuvir RASs are present in Pakistani HCV GT-3a isolates, and they should be monitored carefully, especially in treatment-experienced patients, for further selection of treatment regimens. SIGNIFICANCE AND IMPACT OF STUDY: HCV RASs have been studied very well across the world but there is scarcity of data regarding this topic in Pakistani population, this study provides data regarding the prevalence of these RASs in Pakistani HCV isolates emphasizing the fact that these RASs must be carefully monitored before starting HCV treatment, especially in treatment failure patients.


Subject(s)
Hepatitis C, Chronic , Sofosbuvir , Humans , Sofosbuvir/therapeutic use , Sofosbuvir/pharmacology , Hepacivirus/genetics , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Hepatitis C, Chronic/drug therapy , Drug Resistance, Viral/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/pharmacology , Viral Nonstructural Proteins/therapeutic use , Genotype , RNA
5.
BMC Gastroenterol ; 21(1): 443, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34819046

ABSTRACT

BACKGROUND: NS5A and NS5B proteins of hepatitis C virus (HCV) are the main targets of compounds that directly inhibit HCV infections. However, the emergence of resistance-associated substitutions (RASs) may cause substantial reductions in susceptibility to inhibitors. METHODS: Viral load and genotyping were determined in eighty-seven naïve HCV-infected patients, and the amplified NS5A and NS5B regions were sequenced by Sanger sequencing. In addition, physicochemical properties, structural features, immune epitopes, and inhibitors-protein interactions of sequences were analyzed using several bioinformatics tools. RESULTS: Several amino acid residue changes were found in NS5A and NS5B proteins; however, we did not find any mutations related to resistance to the treatment in NS5B. Different phosphorylation and few glycosylation sites were assessed. Disulfide bonds were identified in both proteins that had a significant effect on the function and structure of HCV proteins. Applying reliable software to predict B-cell epitopes, 3 and 5 regions were found for NS5A and NS5B, respectively, representing a considerable potential to induce the humoral immune system. Docking analysis determined amino acids involved in the interaction of inhibitors and mentioned proteins may not decrease the drug efficiency. CONCLUSIONS: Strong interactions between inhibitors, NS5A and NS5B proteins and the lack of efficient drug resistance mutations in the analyzed sequences may confirm the remarkable ability of NS5A and NS5B inhibitors to control HCV infection amongst Iranian patients. The results of bioinformatics analysis could unveil all features of both proteins, which can be beneficial for further investigations on HCV drug resistance and designing novel vaccines.


Subject(s)
Hepatitis C, Chronic , Hepatitis C , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Resistance, Viral/genetics , Genotype , Hepacivirus/genetics , Hepatitis C/drug therapy , Hepatitis C, Chronic/drug therapy , Humans , Iran , Molecular Docking Simulation , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/pharmacology , Viral Nonstructural Proteins/therapeutic use
6.
Antiviral Res ; 194: 105164, 2021 10.
Article in English | MEDLINE | ID: mdl-34411654

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is an acute infectious disease caused by a novel phlebovirus (SFTSV), characterized by fever, thrombocytopenia and leukocytopenia which lead to multiple organ failure with high mortality in severe cases. The SFTSV has spread rapidly in recent years and posed a serious threat to public health in endemic areas. However, specific antiviral therapeutics for SFTSV infection are rare. In this study, we demonstrated that two peptides, SGc1 and SGc8, derived from a hydrophobic region of the SFTSV glycoprotein Gc, could potently inhibit SFTSV replication in a dose-dependent manner without apparent cytotoxicity in various cell lines and with low immunogenicity and good stability. The IC50 (50% inhibition concentration) values for both peptides to inhibit 2 MOI of SFTSV infection were below 10 µM in L02, Vero and BHK21 cells. Mechanistically, SGc1 and SGc8 mainly inhibited viral entry at the early stage of the viral infection. Inhibition of SFTSV replication was specific by both peptides because no inhibitory effect was shown against other viruses including Zika virus and Enterovirus A71. Taken together, our results suggested that viral glycoprotein-derived SGc1 and SGc8 peptides have antiviral potential and warrant further assessment as an SFTSV-specific therapeutic.


Subject(s)
Antiviral Agents/pharmacology , Glycoproteins/pharmacology , Peptides/pharmacology , Phlebovirus/chemistry , Phlebovirus/drug effects , Viral Nonstructural Proteins/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Enterovirus A, Human/drug effects , Female , Glycoproteins/chemistry , Inhibitory Concentration 50 , Mice , Peptides/chemistry , Phlebovirus/genetics , Severe Fever with Thrombocytopenia Syndrome/drug therapy , Vero Cells , Virus Internalization/drug effects , Virus Replication/drug effects , Zika Virus/drug effects
7.
Curr HIV Res ; 19(5): 391-397, 2021.
Article in English | MEDLINE | ID: mdl-34238162

ABSTRACT

BACKGROUND: Hepatitis C virus (HCV) acts in the host as a complicated mixture of related variants with the potency to genetically escape host immune responses. Direct acting antivirals (DAAs) have been approved for HCV treatment with shorter duration, better cure rates and lower side effects. However, naturally occurring resistance associated substitutions (RASs) create some obstacles to this antiviral therapy success. OBJECTIVE: In this study, we aimed at the determination of the naturally occurring NS3/4A RASs in HCV/human immunodeficiency virus (HIV)infected patients. METHODS: A total of 120 DAA-naïve HCV-HIV co-infected patients were included. HCV NS3/4Agenome region was amplified with PCR and mutation analysis was performed by Sanger sequencing technique. The amino acid sequence diversity of the region was analyzed using geno2pheno HCV. RESULTS: Phylogenetic analysis showed that 73 cases were infected by 3a and 47 subjects by subtype1a. The overall RASs among studied subjects were observed in 6 (5%) individuals from 120 studied cases who were infected with HCV 1a. V36M/L, Q80L, S122G/L, R155T/G, A156S, D168Y/N and S174A/N/T mutations were detected in this study. CONCLUSION: Although the prevalence of RASs was totally low in this study, the presence of several cases of double and triple mutants among this population suggests prior evaluation of protease inhibitors related mutations before initiation of standard treatment and also an investigation on a large population could be of high value.


Subject(s)
HIV Infections , Hepatitis C, Chronic , Hepatitis C , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Resistance, Viral/genetics , Genotype , HIV Infections/drug therapy , HIV Infections/epidemiology , Hepacivirus/genetics , Hepatitis C/drug therapy , Hepatitis C/epidemiology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/epidemiology , Humans , Iran/epidemiology , Phylogeny , Prevalence , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/pharmacology
8.
Res Vet Sci ; 137: 68-76, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33933710

ABSTRACT

In this study, the effects of Goose parvovirus (GPV) infection as well as the possible role of NS1 protein on apoptosis induction in goose embryo fibroblast (GEF) cells were examined. Flow cytometry analysis and TUNEL assays revealed that GPV infection and NS1 transfection induced significant apoptosis in GEF cells compared to what was observed in mock-infected cells. Interestingly, the increase in the rate of apoptosis detected in GPV-infected GEFs was accompanied by an increased viral load in the cells. In addition, the apoptotic pathway was mediated by apoptosis-inducing factors (AIFs) and internal factors that influence the release of AIFs. The results indicated that the mitochondrial membrane potential was decreased, and AIF expression was increased in the nucleus (P < 0.01). Reactive oxygen species (ROS) increased gradually within 48 h (P < 0.001). Cathepsin D activities were also increased (P < 0.05). The results demonstrated that the AIF-mediated pathway is a new mitochondrial apoptotic pathway and that mitochondrial depolarization, ROS content, and cathepsin D activities are the key factors influencing apoptosis in GEF cells.


Subject(s)
Fibroblasts/virology , Geese/embryology , Parvovirinae/pathogenicity , Viral Nonstructural Proteins/pharmacology , Animals , Apoptosis , Apoptosis Inducing Factor/metabolism , Apoptosis Inducing Factor/pharmacology , Cathepsin D/metabolism , Cell Death , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
9.
J Hepatol ; 75(4): 820-828, 2021 10.
Article in English | MEDLINE | ID: mdl-34023351

ABSTRACT

BACKGROUND & AIMS: Retreatment with glecaprevir/pibrentasvir (G/P) resulted in a rate of sustained virologic response 12 weeks after treatment completion (SVR12) of >90% in HCV genotype 1 (GT1) patients who previously failed a regimen of sofosbuvir plus an NS5A inhibitor (NS5Ai). This study investigated the prevalence and impact of baseline NS3 and NS5A resistance-associated substitutions (RASs) on the efficacy of G/P in prior GT1 sofosbuvir+NS5Ai failures and the persistence of treatment-emergent RASs. METHODS: Longitudinal samples from 177 patients enrolled in a phase IIIb, randomized pragmatic clinical trial were analyzed. Patients without cirrhosis were randomized to 12 or 16 weeks of G/P, and patients with compensated cirrhosis were randomized to G/P and ribavirin for 12 weeks or G/P for 16 weeks. Linkage of RAS was identified using Primer-ID next-generation sequencing at a 15% cut-off. RESULTS: Of 177 patients, 169 (95.5%) were PI-naïve. All 33 GT1b-infected patients achieved SVR12. In GT1a-infected patients, baseline NS5A RASs were prevalent (74.5%, 105/141) but NS3 RASs were uncommon. Baseline NS3 RASs had no impact on G/P efficacy and patients with baseline NS5A RASs showed a numerically but not statistically significantly lower SVR12 rate compared to those without NS5A RASs (89% vs. 97%). SVR12 was achieved in 34 of 35 (97%) patients without NS5A baseline substitution, and 53 of 57 (93%), 35 of 40 (88%), 5 of 8 (63%) with single, double-linked, and triple-linked NS5A substitutions, respectively. Among 13 patients with virologic failure, 4 acquired treatment-emergent NS3 RASs and 10 acquired NS5A RASs. CONCLUSION: Baseline NS5A RASs were highly prevalent. The presence of an increasing number of linked NS5A RASs in GT1a showed a trend in decreasing SVR12 rates, although no specific NS5A RASs or their linkage pattern were associated with lower SVR12 rates. LAY SUMMARY: Direct-acting antivirals have revolutionized the treatment of chronic hepatitis C infection, but treatment failure occurs in some patients. Retreatment of patients who previously failed a regimen consisting of sofosbuvir and an NS5A inhibitor with a regimen of glecaprevir and pibrentasvir (G/P) is >90% effective. Herein, we analyzed samples from these patients and showed that retreatment efficacy with G/P is lower in patients with double- or triple-linked NS5A resistance mutations than in patients with single or no NS5A resistance mutations. CLINICAL TRIAL NUMBER: NCT03092375.


Subject(s)
Benzimidazoles/pharmacology , Drug Resistance/immunology , Pyrrolidines/pharmacology , Quinoxalines/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Sofosbuvir/metabolism , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , Benzimidazoles/therapeutic use , Drug Combinations , Female , Hepatitis C/drug therapy , Hepatitis C/epidemiology , Hepatitis C/physiopathology , Humans , Longitudinal Studies , Male , Middle Aged , Pyrrolidines/therapeutic use , Quinoxalines/administration & dosage , Quinoxalines/therapeutic use , RNA-Dependent RNA Polymerase/pharmacology , Sofosbuvir/administration & dosage , Sulfonamides/therapeutic use , United States/epidemiology , Viral Nonstructural Proteins/pharmacology
10.
Protein Expr Purif ; 184: 105892, 2021 08.
Article in English | MEDLINE | ID: mdl-33895264

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is newly discovered virus which is the member of the order Bunyavirales, family phenuiviridae, phlebovirus genus. Its genome is composed of 3 segments of negative-sense RNA L, M and S. NSs is a non structure protein encoded by S segment which is important for viral replication and virulence. NSs protein of SFTSV is only involved in the regulation of host innate immune responses and suppression of IFN-promoter activities. So, the exact functions of this protein need to be studied deeply. To understand the exact role of NSs from SFTSV in viral replication and host immune response, a qualified antibody against this protein is required. In this study, NSs gene of SFTSV, was cloned into a bacterial expression vector (pGEX-6P-1) and the recombinant plasmid was transformed into Escherichia coli BL21 (DE3) cells. The SFTSV NSs fusion protein was purified using Glutathione Sepharose 4B and utilized as an antigen to immunize rabbits and obtain an anti-SFTSV NSs polyclonal antibody. Proper expression of the fusion protein and polyclonal antibody specificity were confirmed by western blotting and immunofluorescence analyses. The polyclonal antibody recognized NSs from SFTSV specifically. This is the first report that NSs can form viroplasm-like structures not only in infected cells but also in transfected cells with NSs plasmids. This polyclonal antibody will be useful for future studies of NSs functions.


Subject(s)
Antibodies, Viral/immunology , Phlebovirus , Viral Nonstructural Proteins , Animals , Chlorocebus aethiops , Humans , Phlebovirus/chemistry , Phlebovirus/genetics , Phlebovirus/immunology , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Vero Cells , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/pharmacology
11.
Cells ; 10(2)2021 01 28.
Article in English | MEDLINE | ID: mdl-33525607

ABSTRACT

Avian reovirus p17 (ARV p17) is a non-structural protein known to activate autophagy, interfere with gene transcription and induce a significant tumor cell growth inhibition in vitro and in vivo. In this study, we show that ARV p17 is capable of exerting potent antiangiogenic properties. The viral protein significantly inhibited the physiological angiogenesis of human endothelial cells (ECs) by affecting migration, capillary-like structure and new vessel formation. ARV p17 was not only able to suppress the EC physiological angiogenesis but also rendered ECs insensitive to two different potent proangiogenic inducers, such as VEGF-A and FGF-2 in the three-dimensional (3D) Matrigel and spheroid assay. ARV p17 was found to exert its antiangiogenic activity by upregulating transcription and release of the well-known tumor suppressor molecule dipeptidyl peptidase 4 (DPP4). The ability of ARV p17 to impact on angiogenesis is completely new and highlights the "two compartments" activity of the viral protein that is expected to hamper the tumor parenchymal/stromal crosstalk. The complex antitumor activities of ARV p17 open the way to a new promising field of research aimed to develop new therapeutic approaches for treating tumor and cancer metastasis.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Neovascularization, Pathologic/metabolism , Orthoreovirus, Avian/metabolism , Viral Nonstructural Proteins/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Movement/drug effects , Chickens , Fibroblast Growth Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Microvessels/cytology , Morphogenesis/drug effects , Recombinant Proteins/pharmacology , Transduction, Genetic , Up-Regulation/drug effects
12.
Biochimie ; 176: 52-61, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32640279

ABSTRACT

Zika virus (ZIKV) infection leads to microcephaly in newborns. Flaviviruses are known to secrete NS1 protein extracellularly and its concentration in serum directly co-relate to disease severity. The presence of ZIKV-NS1 near the brain microvascular endothelial cells (BMVECs) affects blood-brain-barrier, which is composed of tight junctions (TJs) and adherens junctions (AJs). Viruses utilize different strategies to circumvent this barrier to enter in brain. The present study demonstrated the mechanism of junctional integrity disruption in BMVECs by ZIKV-NS1 protein exposure. The Transendothelial Electrical Resistance and sodium fluorescein migration assays revealed the endothelial barrier disruption in BMVECs exposed to ZIKV-NS1 at different time (12hr and 24hr) and doses (500 ng/mL, 1000 ng/mL and 1500 ng/mL). The exposure of ZIKV-NS1 on BMVECs led to the phosphorylation of AJs and suppression of TJs through secreted ZIKV-NS1 in a bystander fashion. The activation of NADPH dependent reactive oxygen species activity and redox sensitive tyrosine kinase further increased the phosphorylation of AJs. The reduced expression of the phosphatase led to the increased phosphorylation of the AJs. The treatment with Diphenyleneiodonium chloride rescued the phosphatase and TJs expression and suppressed the expression of kinase and AJs in BMVECs exposed to ZIKV-NS1.


Subject(s)
Adherens Junctions/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Microvessels/metabolism , Tight Junctions/metabolism , Viral Nonstructural Proteins/metabolism , Zika Virus/metabolism , Humans , Viral Nonstructural Proteins/pharmacology
13.
Biochem Biophys Res Commun ; 516(2): 515-520, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31230744

ABSTRACT

Glioma is the most common primary brain tumor with high mortality. Given the poor outcomes with standard-of-care treatments, novel treatment strategies are needed. Oncolytic viral therapy for glioma has developed as an exciting therapeutic method in recent years. Zika virus, a member of flavivirus family, has oncolytic activity against glioma cells but the mechanism is unknown. Here, we aimed to determine which viral protein might play a critical role in mitigating glioma cell growth. We examined the tumor suppressor function of four nonstructural proteins NS1, NS3, NS4B and NS5 in human glioma cell line U87. As a result, we found that only NS5 significantly inhibited proliferation, migration and invasion of U87 cells. Moreover, expression of NS5 suppressed tumorigenicity of mouse GL261 glioma cell in vivo. Our findings provide some clues for further exploration of oncolytic Zika virus in the treatment of glioma.


Subject(s)
Glioma/pathology , Viral Nonstructural Proteins/pharmacology , Zika Virus/metabolism , Animals , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Neoplasm Invasiveness , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology
14.
Int Immunopharmacol ; 73: 270-279, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31125926

ABSTRACT

BACKGROUND: Dengue fever is a re-emerging viral disease and affects millions of population worldwide. Monocytes are involved in dengue viral disease, however, their exact role is still not clear. In the present study, we investigated, the effect of NS1 antigen of dengue virus and paracetamol on THP-1 monocytes associated to expressions of matrix metalloproteinases (MMPs) and cytokine release. METHODS: Assessment of cell morphology by bright field microscopy, cell viability by MTT assay, protein estimation by Bradford reagent were done in cells exposed to NS1 antigen in the presence and absence of paracetamol. Cytokines estimations were done by ELISA. Expression profile of matrix metalloproteinase genes was done using real-time PCR and reverse-transcriptase PCR. RESULTS: NS1 exposure of THP-1 monocytes cells, changed their cell morphology and activated them for release of proteins in 24 h. Expressions of MMP-2, MMP-8, MMP-9 and MMP-14 genes were upregulated by NS1 exposure. Further, exposure of NS1 to THP-1 monocytes cells increased expression profile of MMP-10 and MMP-13 genes to a lesser extent. Treatment with paracetamol (1 mg/ml and 2 mg/ml), significantly down-regulated the expression profile of MMP-2, MMP-8, MMP-9 and 14 in dose dependent manner. NS1 exposure also increased the release of cytokines IL-4, IL-6, and IL-10 but decreased the release of TNF-α and IL-15. Interestingly, paracetamol reversed NS1 induced changes in the release of these cytokine in dose dependent manner. CONCLUSION: Monocytes mediated expression of MMPs participates in the development of dengue pathogenesis in the severe cases of disease and paracetamol may have a protective effect in dengue viral disease.


Subject(s)
Acetaminophen/pharmacology , Matrix Metalloproteinases/genetics , Monocytes/drug effects , Protective Agents/pharmacology , Viral Nonstructural Proteins/pharmacology , Cytokines/metabolism , Dengue , Humans , Monocytes/metabolism , RNA, Messenger/metabolism , THP-1 Cells
15.
Arch Virol ; 164(1): 255-260, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30259142

ABSTRACT

Bovine viral diarrhea virus (BVDV) is an important pathogen in cattle that causes economic losses in livestock industries. Autophagy is an essential cell system for the maintenance of homeostasis and is induced by various triggers, including infection by viruses. BVDV infection leads to autophagy in order to enhance its replication in cells. In this study, we investigated the effect of BVDV non-structural proteins on the induction of autophagosomes. We found that NS4B alone could induce autophagosomes, suggesting a novel and important function of NS4B in BVDV replication.


Subject(s)
Autophagosomes/drug effects , Autophagy/physiology , Diarrhea Viruses, Bovine Viral/metabolism , Kidney/cytology , Viral Nonstructural Proteins/pharmacology , Animals , Autophagosomes/physiology , Cattle , Cell Line , Viral Nonstructural Proteins/metabolism
16.
Viral Immunol ; 31(8): 575-582, 2018 10.
Article in English | MEDLINE | ID: mdl-30281404

ABSTRACT

There is a growing body of studies that show the important role of NS3 protein from hepatitis C virus in fibrosis. However, mechanisms of the effects of this protein on immune modulation of stellate cells remain to be investigated. Herein, the effect of NS3 protein on the expression level of suppressor of cytokine signaling (SOCS)1/3 and interleukin-24 (IL-24)-related genes was investigated in hepatic stellate cell (HSC), LX-2. Recombinant NS3 protein was added to LX-2 HSC culture. Leptin and standard medium treatments were also included in experiments as positive and negative controls, respectively. Total RNA was extracted from each well at 6, 12, and 24 h after NS3 addition. The expression levels of the fibrotic (transforming growth factor beta 1 [TGF-ß], alpha-smooth muscle actin [α-SMA], and COL1A1), inflammatory (IL-6 and IL-24), IL-20R, IL-22R, and immunosuppressive genes (SOCS1 and SOCS3) were evaluated by real-time polymerase chain reaction (PCR). Recombinant NS3 protein induced activated phenotypes of LX-2 with a significant increase in the expression level of α-SMA COL1A1 (p < 0.0001) and TGF-ß. Moreover, this exposure led to a meaningful elevation in the expression of IL-6. Furthermore, compared with leptin (control), after the stellate cell treatment with NS3, SOCS1 and SOCS3 gene expression induced at a comparable level. Compared with the control sample, the NS3 protein significantly increased the expression level of IL-24 and its related receptors, IL-20R and IL-22R. This study not only confirmed the previously proved inflammatory and fibrotic effect of this protein but also indicated that high expression levels of SOCS1, SOCS3, and IL-24 have a significant effect on HSC activation. Therefore, these two molecules can be used as a potential therapeutic target candidate.


Subject(s)
Gene Expression/immunology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/immunology , Immunologic Factors/genetics , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/pharmacology , Cells, Cultured , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Interleukins/genetics , Interleukins/immunology , Leptin/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Real-Time Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/immunology , Viral Nonstructural Proteins/genetics
17.
J Proteome Res ; 17(4): 1474-1484, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29558158

ABSTRACT

Influenza A virus infections can result in severe respiratory diseases. The H7N9 subtype of avian influenza A virus has been transmitted to humans and caused severe disease and death. Nonstructural protein 1 (NS1) of influenza A virus is a virulence determinant during viral infection. To elucidate the functions of the NS1 encoded by influenza A H7N9 virus (H7N9 NS1), interaction partners of H7N9 NS1 in human cells were identified with immunoprecipitation followed by SDS-PAGE coupled with liquid chromatography-tandem mass spectrometry (GeLC-MS/MS). We identified 36 cellular proteins as the interacting partners of the H7N9 NS1, and they are involved in RNA processing, mRNA splicing via spliceosome, and the mRNA surveillance pathway. Two of the interacting partners, cleavage and polyadenylation specificity factor subunit 2 (CPSF2) and CPSF7, were confirmed to interact with H7N9 NS1 using coimmunoprecipitation and immunoblotting based on the previous finding that the two proteins are involved in pre-mRNA polyadenylation machinery. Furthermore, we illustrate that overexpression of H7N9 NS1, as well as infection by the influenza A H7N9 virus, interfered with pre-mRNA polyadenylation in host cells. This study comprehensively profiled the interactome of H7N9 NS1 in host cells, and the results demonstrate a novel endotype for H7N9 NS1 in inhibiting host mRNA maturation.


Subject(s)
Influenza A Virus, H7N9 Subtype/chemistry , RNA, Messenger/antagonists & inhibitors , Viral Nonstructural Proteins/pharmacology , Animals , Cleavage And Polyadenylation Specificity Factor , Host Microbial Interactions , Humans , Immunoblotting , Immunoprecipitation , Influenza A Virus, H7N9 Subtype/pathogenicity , Protein Binding , mRNA Cleavage and Polyadenylation Factors
18.
Article in English | MEDLINE | ID: mdl-29406280

ABSTRACT

The red-legged partridge (Alectoris rufa) is a competent host for West Nile virus (WNV) replication and highly susceptible to WNV disease. With the aim to assess in this species whether the inoculation of non-structural protein NS1 from WNV elicits a protective immune response against WNV infection, groups of partridges were inoculated with recombinant NS1 (NS1 group) or an unrelated recombinant protein (mock group), and challenged with infectious WNV. A third group received no inoculation prior to challenge (challenge group). The NS1 group failed to elicit detectable antibodies to NS1 while in the mock group a specific antibody response was observed. Moreover, no protection against WNV disease was observed in the NS1 group, but rather, it showed significantly higher viral RNA load and delayed neutralizing antibody response, and suffered a more severe clinical disease, which resulted in higher mortality. This adverse effect has not been observed before and warrants further investigations.


Subject(s)
Bird Diseases/virology , Galliformes/virology , Viral Nonstructural Proteins/pharmacology , West Nile Fever/veterinary , West Nile virus/immunology , Animals , Bird Diseases/immunology , Bird Diseases/prevention & control , Galliformes/immunology , Immunity, Humoral/drug effects , Recombinant Proteins , Viral Nonstructural Proteins/immunology , West Nile Fever/immunology , West Nile Fever/prevention & control
19.
BMC Microbiol ; 17(1): 124, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28545480

ABSTRACT

BACKGROUND: Hepatitis C virus (HCV) core protein and nonstructural protein 4B (NS4B) are potentially oncogenic. Aberrant activation of the Wnt/ß-catenin signaling pathway is closely associated with hepatocarcinogenesis. We investigated the effects of HCV type 1b core protein and NS4B on Wnt/ß-catenin signaling in various liver cells, and explored the molecular mechanism underlying HCV-related hepatocarcinogenesis. RESULTS: Compared with the empty vector control, HCV core protein and NS4B demonstrated the following characteristics in the Huh7 cells: significantly enhanced ß-catenin/Tcf-dependent transcriptional activity (F = 40.87, P < 0.01); increased nuclear translocation of ß-catenin (F = 165.26, P < 0.01); upregulated nuclear ß-catenin, cytoplasmic ß-catenin, Wnt1, c-myc, and cyclin D1 protein expression (P < 0.01); and promoted proliferation of Huh7 cells (P < 0.01 or P < 0.05). Neither protein enhanced ß-catenin/Tcf-dependent transcriptional activity in the LO2 cells (F = 0.65, P > 0.05), but they did significantly enhance Wnt3a-induced ß-catenin/Tcf-dependent transcriptional activity (F = 64.25, P < 0.01), and promoted the nuclear translocation of ß-catenin (F = 66.54, P < 0.01) and the Wnt3a-induced proliferation of LO2 cells (P < 0.01 or P < 0.05). Moreover, activation of the Wnt/ß-catenin signaling pathway was greater with the core protein than with NS4B (P < 0.01 or P < 0.05). CONCLUSIONS: HCV core protein and NS4B directly activate the Wnt/ß-catenin signaling pathway in Huh7 cells and LO2 cells induced by Wnt3a. These data suggest that HCV core protein and NS4B contribute to HCV-associated hepatocellular carcinogenesis.


Subject(s)
Hepatitis C/metabolism , Viral Core Proteins/pharmacology , Viral Nonstructural Proteins/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/drug effects , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Humans , Mice , Signal Transduction/drug effects , Transcription, Genetic , beta Catenin/metabolism
20.
Viruses ; 9(3)2017 03 13.
Article in English | MEDLINE | ID: mdl-28335410

ABSTRACT

The Flaviviridae family comprises a number of human pathogens, which, although sharing structural and functional features, cause diseases with very different outcomes. This can be explained by the plurality of functions exerted by the few proteins coded by viral genomes, with some of these functions shared among members of a same family, but others being unique for each virus species. These non-canonical functions probably have evolved independently and may serve as the base to the development of specific therapies for each of those diseases. Here it is discussed what is currently known about the non-canonical roles of dengue virus (DENV) non-structural proteins (NSPs), which may account for some of the effects specifically observed in DENV infection, but not in other members of the Flaviviridae family. This review explores how DENV NSPs contributes to the physiopathology of dengue, evasion from host immunity, metabolic changes, and redistribution of cellular components during infection.


Subject(s)
Dengue Virus/physiology , Dengue Virus/pathogenicity , Host-Pathogen Interactions , Viral Nonstructural Proteins/pharmacology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...