Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
2.
J Virol ; 97(11): e0096323, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37846984

ABSTRACT

IMPORTANCE: Currently licensed dengue vaccines do not induce long-term protection in children without previous exposure to dengue viruses in nature. These vaccines are based on selected attenuated strains of the four dengue serotypes and employed in combination for two or three consecutive doses. In our search for a better dengue vaccine candidate, live attenuated strains were followed by non-infectious virus-like particles or the plasmids that generate these particles upon injection into the body. This heterologous prime-boost immunization induced elevated levels of virus-specific antibodies and helped to prevent dengue virus infection in a high proportion of vaccinated macaques. In macaques that remained susceptible to dengue virus, distinct mechanisms were found to account for the immunization failures, providing a better understanding of vaccine actions. Additional studies in humans in the future may help to establish whether this combination approach represents a more effective means of preventing dengue by vaccination.


Subject(s)
Dengue Vaccines , Dengue Virus , Dengue , Vaccines, Virus-Like Particle , Animals , Humans , Antibodies, Viral , Dengue Vaccines/administration & dosage , Macaca fascicularis , Immunization, Secondary , Vaccines, Virus-Like Particle/administration & dosage
3.
Viruses ; 13(7)2021 07 17.
Article in English | MEDLINE | ID: mdl-34372598

ABSTRACT

Dengue virus (DENV) infection causes a spectrum of dengue diseases that have unclear underlying mechanisms. Nonstructural protein 1 (NS1) is a multifunctional protein of DENV that is involved in DENV infection and dengue pathogenesis. This study investigated the potential post-translational modification of DENV NS1 by phosphorylation following DENV infection. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), 24 potential phosphorylation sites were identified in both cell-associated and extracellular NS1 proteins from three different cell lines infected with DENV. Cell-free kinase assays also demonstrated kinase activity in purified preparations of DENV NS1 proteins. Further studies were conducted to determine the roles of specific phosphorylation sites on NS1 proteins by site-directed mutagenesis with alanine substitution. The T27A and Y32A mutations had a deleterious effect on DENV infectivity. The T29A, T230A, and S233A mutations significantly decreased the production of infectious DENV but did not affect relative levels of intracellular DENV NS1 expression or NS1 secretion. Only the T230A mutation led to a significant reduction of detectable DENV NS1 dimers in virus-infected cells; however, none of the mutations interfered with DENV NS1 oligomeric formation. These findings highlight the importance of DENV NS1 phosphorylation that may pave the way for future target-specific antiviral drug design.


Subject(s)
Dengue Virus/chemistry , Dengue Virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Chromatography, Liquid , Dengue/virology , Dengue Virus/genetics , Hep G2 Cells , Humans , Kinetics , Phosphorylation , Protein Binding , Sequence Analysis, Protein , Tandem Mass Spectrometry , Vero Cells , Viral Nonstructural Proteins/genetics , Virus Replication
4.
Viruses ; 13(4)2021 04 11.
Article in English | MEDLINE | ID: mdl-33920458

ABSTRACT

Dengue virus (DENV) is the causative pathogen in the life-threatening dengue hemorrhagic fever and dengue shock syndrome. DENV is transmitted to humans via the bite of an infected Aedes mosquito. Approximately 100 million people are infected annually worldwide, and most of those live in tropical and subtropical areas. There is still no effective drug or vaccine for treatment of DENV infection. In this study, we set forth to investigate the effect of melatonin, which is a natural hormone with multiple pharmacological functions, against DENV infection. Treatment with subtoxic doses of melatonin dose-dependently inhibited DENV production. Cross-protection across serotypes and various cell types was also observed. Time-of-addition assay suggested that melatonin exerts its influence during the post-entry step of viral infection. The antiviral activity of melatonin partly originates from activation of the sirtuin pathway since co-treatment with melatonin and the sirtuin 1 (SIRT1) inhibitor reversed the effect of melatonin treatment alone. Moreover, melatonin could modulate the transcription of antiviral genes that aid in suppression of DENV production. This antiviral mechanism of melatonin suggests a possible new strategy for treating DENV infection.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Interferons/immunology , Melatonin/pharmacology , Metabolic Networks and Pathways/drug effects , Sirtuin 1/metabolism , Virus Replication/drug effects , A549 Cells , Aedes , Animals , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Dengue/drug therapy , Humans , Metabolic Networks and Pathways/immunology , Vero Cells
5.
Sci Rep ; 10(1): 12933, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737386

ABSTRACT

Viruses manipulate the life cycle in host cells via the use of viral properties and host machineries. Development of antiviral peptides against dengue virus (DENV) infection has previously been concentrated on blocking the actions of viral structural proteins and enzymes in virus entry and viral RNA processing in host cells. In this study, we proposed DENV NS1, which is a multifunctional non-structural protein indispensable for virus production, as a new target for inhibition of DENV infection by specific peptides. We performed biopanning assays using a phage-displayed peptide library and identified 11 different sequences of 12-mer peptides binding to DENV NS1. In silico analyses of peptide-protein interactions revealed 4 peptides most likely to bind to DENV NS1 at specific positions and their association was analysed by surface plasmon resonance. Treatment of Huh7 cells with these 4 peptides conjugated with N-terminal fluorescent tag and C-terminal cell penetrating tag at varying time-of-addition post-DENV infection could inhibit the production of DENV-2 in a time- and dose-dependent manner. The inhibitory effects of the peptides were also observed in other virus serotypes (DENV-1 and DENV-4), but not in DENV-3. These findings indicate the potential application of peptides targeting DENV NS1 as antiviral agents against DENV infection.


Subject(s)
Antiviral Agents , Dengue Virus/physiology , Dengue , Drug Delivery Systems , Peptide Library , Viral Nonstructural Proteins , Virus Replication/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line, Tumor , Dengue/drug therapy , Dengue/metabolism , Humans , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
6.
Virus Genes ; 56(1): 27-36, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31720911

ABSTRACT

Dengue virus (DENV) requires clathrin-mediated endocytosis for its entry into the cells where the adaptor protein complex (AP) is vital for the clathrin-coated vesicle formation. The role of AP-2 was previously examined in the early stages of DENV infection; however, the role of AP-2 in the late stage of DENV infection was not determined. The µ1 subunit of AP-2 (AP2M1) is one of the most important cytoplasmic carrier domains in clathrin-mediated endocytosis and the phosphorylation of this subunit by the kinase enzyme, AP-2 associated protein kinase 1 (AAK1), stimulates clathrin and supports the cell surface receptor incorporation. In the present study, we primarily aimed to investigate the role of AP2M1 by gene silencing approach as well as using naked DENV RNA transfection into AP2M1 knockdown cells. Secondarily, an inhibitor of AAK1, sunitinib was used to investigate whether AAK1 could influence the virus production in DENV-infected Huh7 cells. The knockdown of AP2M1 in the DENV-infected Huh7 cells displayed a reduction in the viral titer at 24 h post-infection. Furthermore, experiments were conducted to bypass the DENV internalization using a naked DENV RNA transfection into the AP2M1 knockdown cells. Higher intracellular DENV RNA, DENV E protein, and intracellular virion were observed, whereas the extracellular virion production was comparably less than that of control. Treatment with sunitinib in DENV-infected Huh7 cells was able to reduce extracellular virion production and was consistent with all four serotypes of DENV. Therefore, our findings demonstrate the role of AP2M1 in the exocytosis step of DENV replication leading to infectious DENV production and the efficacy of sunitinib in suppressing virus production during the infection with different serotypes of DENV.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Dengue Virus/physiology , Dengue/virology , Virus Release , Adaptor Proteins, Vesicular Transport/genetics , Cell Line , Dengue/physiopathology , Dengue Virus/genetics , Endocytosis , Host-Pathogen Interactions , Humans , Virus Replication
7.
Virus Res ; 271: 197672, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31386864

ABSTRACT

Dengue virus (DENV) infection has evolved into a major global health menace and economic burden due to its intensity and geographic distribution. DENV infection in humans can cause a wide range of symptoms including dengue fever (DF), dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). An antiviral agent that is effective against all four serotypes of DENV is urgently needed to prevent and to manage this condition. Reducing the viral load during the early phase of infection may minimize the chance of patients progressing to more severe DHF or DSS. In this study, we set forth to investigate the anti-viral effect of five commercially available protease inhibitors on DENV infection since both viral and host proteases can contribute to effective viral replication. Previously, the serine protease inhibitor AEBSF [4-(2-aminoethyl) benzene sulfonyl fluoride] has been shown to inhibit DENV NS3 protease activity. The results of the present study revealed that DENV genome replication and protein synthesis were significantly inhibited by AEBSF in a dose-dependent manner. AEBSF inhibited the expression of genes such as 3-hydroxy 3-methyl-glutaryl-CoA synthase (HMGCS), 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR), and low-density lipoprotein receptor (LDLR). Moreover, AEBSF significantly inhibited HMGCR activity and intracellular cholesterol synthesis after DENV infection. The anti-DENV effect of AEBSF was confirmed in all four DENV serotypes and in three different cell lines. These results indicate that AEBSF reduces DENV infection via both viral and host protease activities.


Subject(s)
Cholesterol/biosynthesis , Dengue Virus/drug effects , Dengue/metabolism , Dengue/virology , Serine Proteinase Inhibitors/pharmacology , Sulfones/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Dengue Virus/classification , Dengue Virus/genetics , Genome, Viral , Humans , Virus Replication
8.
Virus Res ; 260: 123-128, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30503719

ABSTRACT

Skin dendritic cells (DCs) are primary target cells of dengue virus (DENV) infection and they play an important role in its immunopathogenesis. Monocyte-derived dendritic cells (MDDCs) represent dermal and bloodstream DCs that serve as human primary cells for ex vivo studies of DENV infection. Improved understanding of the mechanisms that effectuate the inhibition of DENV replication in MDDCs will accelerate the development of antiviral drugs to treat DENV infection. In this study, we investigated whether or not vivo-morpholino oligomer (vivo-MO), which was designed to target the top of the 3' stem-loop (3' SL) at the 3' UTR of the DENV genome, could inhibit DENV infection and replication in MDDCs. The findings of this study revealed that vivo-MO-1 could inhibit DENV-2 infection in MDDCs, and that it could significantly reduce DENV RNA, protein, and viral production in a dose-dependent manner. Treatment of MDDCs with 4 µM of vivo-MO-1 decreased DENV production by more than 1,000-fold, when compared to that of the vivo-MO-NC control. Thus, vivo-MO-1 targeting of DENV RNA demonstrates potential for further development into an anti-DENV agent.


Subject(s)
Antiviral Agents/pharmacology , Dendritic Cells/virology , Dengue Virus/drug effects , Dengue Virus/physiology , Oligonucleotides, Antisense/pharmacology , Virus Replication/drug effects , Cells, Cultured , Humans
9.
J Gen Virol ; 99(10): 1391-1406, 2018 10.
Article in English | MEDLINE | ID: mdl-30102148

ABSTRACT

Virus-host interactions play important roles in virus infection and host cellular response. Several viruses, including dengue virus (DENV), usurp host chaperones to support their amplification and survival in the host cell. We investigated the interaction of nonstructural protein 1 (NS1) of DENV with three endoplasmic reticulum-resident chaperones (i.e. GRP78, calnexin and calreticulin) to delineate their functional roles and potential binding sites for protein complex formation. GRP78 protein showed prominent association with DENV NS1 in virus-infected Huh7 cells as evidenced by co-localization and co-immunoprecipitation assays. Further studies on the functional interaction of GRP78 protein were performed by using siRNA-mediated gene knockdown in a DENV replicon transfection system. GRP78 knockdown significantly decreased intracellular NS1 production and delayed NS1 secretion but had no effect on viral RNA replication. Dissecting the important domain of GRP78 required for DENV NS1 interaction showed co-immunoprecipitation of DENV NS1 with a full-length and substrate-binding domain (SBD), but not an ATPase domain, of GRP78, confirming their interaction through SBD binding. Molecular dynamics simulations of DENV NS1 and human GRP78 complex revealed their potential binding sites through hydrogen and hydrophobic bonding. The majority of GRP78-binding sites were located in a ß-roll domain and connector subdomains on the DENV NS1 structure involved in hydrophobic surface formation. Taken together, our findings demonstrated the roles of human GRP78 in facilitating the intracellular production and secretion of DENV NS1 as well as predicted potential binding sites between the DENV NS1 and GRP78 complex, which could have implications in the future development of target-based antiviral drugs.


Subject(s)
Dengue Virus/growth & development , Heat-Shock Proteins/metabolism , Host-Pathogen Interactions , Viral Nonstructural Proteins/metabolism , Calnexin/metabolism , Calreticulin/metabolism , Cell Line , Endoplasmic Reticulum Chaperone BiP , Hepatocytes/virology , Humans , Immunoprecipitation , Molecular Dynamics Simulation , Protein Binding , Protein Multimerization , Virus Replication
10.
Virus Res ; 255: 171-178, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30055216

ABSTRACT

Dengue virus (DENV) disease outbreaks continue to develop across the globe with significant associated mortality and economic burden, yet no treatment has been approved to combat this virus. In an attempt to identify novel drug candidates as therapeutics for DENV infection, we evaluated four US Food and Drug Administration (FDA) approved drugs including aminolevullic acid, azelaic acid, mitoxantrone hydrochloride, and quinine sulfate, and tested their ability to inhibit DENV replication using focus-forming unit assay to quantify virus production. Of the four investigated compounds, quinine was found to have the most pronounced anti-DENV activity. Quinine inhibited DENV production of DENV by about 80% compared to untreated controls, while the other three drugs decreased virus production by only about 50%. Moreover, quinine inhibited DENV production of all four serotypes of DENV. Reduction in virus production was documented in three different cell lines of human origin. Quinine significantly inhibited DENV replication by reducing DENV RNA and viral protein synthesis in a dose-dependent manner. In addition, quinine ameliorated expression of genes related to innate immune response. These findings suggest the efficacy of quinine for stimulating antiviral genes to reduce DENV replication. The antiviral activity of quinine observed in this study may have applicability in the development of new drug therapies against DENV.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue/virology , Drug Repositioning , Quinine/pharmacology , Animals , Antigens, Viral/analysis , Cell Line , Chlorocebus aethiops , Dengue Virus/physiology , Humans , Immunity, Innate/drug effects , Models, Biological , Serogroup , Vero Cells , Viral Load/drug effects , Virus Replication/drug effects
11.
Virus Res ; 250: 13-20, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29608995

ABSTRACT

Dengue hemorrhagic fever is a life-threatening disease caused by the dengue virus (DENV). After DENV enters into host cells, it replicates to generate viral particles to infect other cells. DENV exploits components of the cellular trafficking pathway to achieve effective virion production. Understanding of the proteins required for this trafficking process is essential for revealing the pathogenesis of DENV infection. Coat protein complex and soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), two host protein families in the cellular trafficking pathway, were investigated to elucidate their respective roles during DENV infection. Coat proteins (COPI and COPII) and SNAREs (STX 5 and NSF) were knocked down in a DENV-infected Huh7 cells by RNA interference. Depletion of COPI and COPII, but not of STX5 and NSF, decreased DENV production in DENV-infected Huh7 cells. DENV proteins, including DENV C, prM, E, and NS1, were significantly reduced in COPI-silenced DENV-infected Huh7 cells, when compared to those of control cells. COPI also facilitated DENV production in an endothelial cell line and in all DENV serotypes, indicating the importance of coat protein complex in facilitating DENV infection.


Subject(s)
Coat Protein Complex I/metabolism , Dengue Virus/physiology , SNARE Proteins/metabolism , Virus Replication , Cell Line , Coat Protein Complex I/genetics , Dengue Virus/pathogenicity , Gene Knockdown Techniques , Host-Pathogen Interactions , Humans , Protein Transport , RNA Interference , SNARE Proteins/genetics , Virion/physiology
12.
Arch Virol ; 163(4): 867-876, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29260328

ABSTRACT

Dengue virus (DENV) infection is a worldwide public health problem, which can cause severe dengue hemorrhagic fever (DHF) and life-threatening dengue shock syndrome (DSS). There are currently no anti-DENV drugs available, and there has been an intensive search for effective anti-DENV agents that can inhibit all four DENV serotypes. In this study, we tested whether vivo-morpholino oligomers (vivo-MOs), whose effect on DENV infection has not previously been studied, can inhibit DENV infection. Vivo-MOs were designed to target the top of 3' stem-loop (3' SL) in the 3' UTR of the DENV genome and tested for inhibition of DENV infection in monkey kidney epithelial (Vero) cells and human lung epithelial carcinoma (A549) cells. The results showed that vivo-MOs could bind to a DENV RNA sequence and markedly reduce DENV-RNA, protein, and virus production in infected Vero and A549 cells. Vivo-MOs at a concentration of 4 µM could inhibit DENV production by more than 104-fold when compared to that of an untreated control. In addition, vivo-MOs also inhibited DENV production in U937 cells and primary human monocytes. Therefore, vivo-MOs targeting to the 3' SL in the 3' UTR of DENV genomes are effective and have the potential to be developed as anti-DENV agents.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Genome, Viral , Morpholinos/genetics , Virus Replication/drug effects , A549 Cells , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Base Pairing , Chlorocebus aethiops , Dengue Virus/genetics , Dengue Virus/growth & development , Humans , Inverted Repeat Sequences/drug effects , Monocytes/drug effects , Monocytes/virology , Morpholinos/metabolism , Nucleic Acid Conformation , Primary Cell Culture , U937 Cells , Vero Cells
13.
PLoS One ; 12(11): e0188121, 2017.
Article in English | MEDLINE | ID: mdl-29145490

ABSTRACT

Hepatic dysfunction is a feature of dengue virus (DENV) infection. Hepatic biopsy specimens obtained from fatal cases of DENV infection show apoptosis, which relates to the pathogenesis of DENV infection. However, how DENV induced liver injury is not fully understood. In this study, we aim to identify the factors that influence cell death by employing an apoptosis-related siRNA library screening. Our results show the effect of 558 gene silencing on caspase 3-mediated apoptosis in DENV-infected Huh7 cells. The majority of genes that contributed to apoptosis were the apoptosis-related kinase enzymes. Tumor necrosis factor superfamily member 12 (TNFSF12), and sphingosine kinase 2 (SPHK2), were selected as the candidate genes to further validate their influences on DENV-induced apoptosis. Transfection of siRNA targeting SPHK2 but not TNFSF12 genes reduced apoptosis determined by Annexin V/PI staining. Knockdown of SPHK2 did not reduce caspase 8 activity; however, did significantly reduce caspase 9 activity, suggesting its involvement of SPHK2 in the intrinsic pathway of apoptosis. Treatment of ABC294649, an inhibitor of SPHK2, reduced the caspase 3 activity, suggesting the involvement of its kinase activity in apoptosis. Knockdown of SPHK2 significantly reduced caspase 3 activity not only in DENV-infected Huh7 cells but also in DENV-infected HepG2 cells. Our results were consistent across all of the four serotypes of DENV infection, which supports the pro-apoptotic role of SPHK2 in DENV-infected liver cells.


Subject(s)
Apoptosis/physiology , Dengue Virus/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , RNA Interference , Caspase 3/metabolism , Cell Line , Gene Knockdown Techniques , Humans , Liver/virology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Real-Time Polymerase Chain Reaction , Virus Replication
14.
Antiviral Res ; 141: 7-18, 2017 05.
Article in English | MEDLINE | ID: mdl-28188818

ABSTRACT

High viral load with liver injury is exhibited in severe dengue virus (DENV) infection. Mitogen activated protein kinases (MAPKs) including ERK1/2 and p38 MAPK were previously found to be involved in the animal models of DENV-induced liver injury. However, the role of JNK1/2 signaling in DENV-induced liver injury has never been investigated. JNK1/2 inhibitor, SP600125, was used to investigate the role of JNK1/2 signaling in the BALB/c mouse model of DENV-induced liver injury. SP600125-treated DENV-infected mice ameliorated leucopenia, thrombocytopenia, hemoconcentration, liver transaminases and liver histopathology. DENV-induced liver injury exhibited induced phosphorylation of JNK1/2, whereas SP600125 reduced this phosphorylation. An apoptotic real-time PCR array profiler was used to screen how SP600125 affects the expression of 84 cell death-associated genes to minimize DENV-induced liver injury. Modulation of caspase-3, caspase-8 and caspase-9 expressions by SP600125 in DENV-infected mice suggests its efficiency in restricting apoptosis via both extrinsic and intrinsic pathways. Reduced expressions of TNF-α and TRAIL are suggestive to modulate the extrinsic apoptotic signals, where reduced p53 phosphorylation and induced anti-apoptotic Bcl-2 expression indicate the involvement of the intrinsic apoptotic pathway. This study thus demonstrates the pivotal role of JNK1/2 signaling in DENV-induced liver injury and how SP600125 modulates this pathogenesis.


Subject(s)
Anthracenes/pharmacology , Liver/pathology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Severe Dengue/metabolism , Severe Dengue/pathology , Animals , Anthracenes/administration & dosage , Anthracenes/therapeutic use , Apoptosis/drug effects , Caspases/drug effects , Dengue Virus/drug effects , Disease Models, Animal , Leukopenia/drug therapy , Liver/drug effects , Liver/virology , Mice , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Phosphorylation/drug effects , Severe Dengue/drug therapy , Severe Dengue/virology , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Necrosis Factor-alpha/genetics , Viral Load , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Biochem Biophys Res Commun ; 483(1): 58-63, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28065855

ABSTRACT

Dengue virus is the causative agent of dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. High rates of dengue virus replication and virion production are related to disease severity. To identify anti-DENV compounds, we performed cell-based ELISA testing to detect the level of DENV E protein expression. Among a total of 83 inhibitors, eight were identified as inhibitors with antiviral activity. Epidermal growth factor receptor inhibitor II (EGFR/ErbB-2/ErbB-4 inhibitor II) and protein tyrosine phosphatase inhibitor IV (PTP inhibitor IV) significantly inhibited dengue virus production and demonstrated low toxicity in hepatocyte cell lines. Our results suggest the efficacy of tyrosine kinase/phosphatase inhibitors in decreasing dengue virus production in HepG2 cells.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Dengue Virus/physiology , Drug Evaluation, Preclinical , ErbB Receptors/antagonists & inhibitors , Hep G2 Cells , Humans , RNA, Viral/biosynthesis , Receptor, ErbB-4/antagonists & inhibitors , Viral Proteins/biosynthesis , Virus Replication/drug effects , Virus Replication/physiology
16.
J Immunol ; 197(10): 4053-4065, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798151

ABSTRACT

Flavivirus nonstructural protein 1 (NS1) is a unique secreted nonstructural glycoprotein. Although it is absent from the flavivirus virion, intracellular and extracellular forms of NS1 have essential roles in viral replication and the pathogenesis of infection. The fate of NS1 in insect cells has been more controversial, with some reports suggesting it is exclusively cell associated. In this study, we confirm NS1 secretion from cells of insect origin and characterize its physical, biochemical, and functional properties in the context of dengue virus (DENV) infection. Unlike mammalian cell-derived NS1, which displays both high mannose and complex type N-linked glycans, soluble NS1 secreted from DENV-infected insect cells contains only high mannose glycans. Insect cell-derived secreted NS1 also has different physical properties, including smaller and more heterogeneous sizes and the formation of less stable NS1 hexamers. Both mammalian and insect cell-derived NS1 bind to complement proteins C1s, C4, and C4-binding protein, as well as to a novel partner, mannose-binding lectin. Binding of NS1 to MBL protects DENV against mannose-binding lectin-mediated neutralization by the lectin pathway of complement activation. As we detected secreted NS1 and DENV together in the saliva of infected Aedes aegypti mosquitoes, these findings suggest a mechanism of viral immune evasion at the very earliest phase of infection.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Dengue Virus/immunology , Immune Evasion , Mannose-Binding Lectin/immunology , Mannose-Binding Lectin/metabolism , Viral Nonstructural Proteins/metabolism , Aedes/virology , Animals , Cell Line , Complement Activation , Complement System Proteins/immunology , Complement System Proteins/metabolism , Dengue Virus/pathogenicity , Humans , Protein Binding , Saliva/virology , Swine , Viral Nonstructural Proteins/chemistry
17.
Biochem Biophys Res Commun ; 478(1): 410-416, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27396621

ABSTRACT

Dengue virus infection is one of the most common arthropod-borne viral diseases. A complex interplay between host and viral factors contributes to the severity of infection. The antiviral effects of three antibiotics, lomefloxacin, netilmicin, and minocycline, were examined in this study, and minocycline was found to be a promising drug. This antiviral effect was confirmed in all four serotypes of the virus. The effects of minocycline at various stages of the viral life cycle, such as during viral RNA synthesis, intracellular envelope protein expression, and the production of infectious virions, were examined and found to be significantly reduced by minocycline treatment. Minocycline also modulated host factors, including the phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2). The transcription of antiviral genes, including 2'-5'-oligoadenylate synthetase 1 (OAS1), 2'-5'-oligoadenylate synthetase 3 (OAS3), and interferon α (IFNA), was upregulated by minocycline treatment. Therefore, the antiviral activity of minocycline may have a potential clinical use against Dengue virus infection.


Subject(s)
Dengue Virus/drug effects , Dengue/drug therapy , Dengue/virology , Drug Repositioning/methods , Minocycline/administration & dosage , Viral Load/drug effects , Antiviral Agents/administration & dosage , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Treatment Outcome , Viral Load/physiology
18.
Biochim Biophys Acta ; 1864(9): 1270-1280, 2016 09.
Article in English | MEDLINE | ID: mdl-27108190

ABSTRACT

Dengue virus (DENV) infection is a leading cause of the mosquito-borne infectious diseases that affect humans worldwide. Virus-host interactions appear to play significant roles in DENV replication and the pathogenesis of DENV infection. Nonstructural protein 1 (NS1) of DENV is likely involved in these processes; however, its associations with host cell proteins in DENV infection remain unclear. In this study, we used a combination of techniques (immunoprecipitation, in-solution trypsin digestion, and LC-MS/MS) to identify the host cell proteins that interact with cell-associated NS1 in an in vitro model of DENV infection in the human hepatocyte HepG2 cell line. Thirty-six novel host cell proteins were identified as potential DENV NS1-interacting partners. A large number of these proteins had characteristic binding or catalytic activities, and were involved in cellular metabolism. Coimmunoprecipitation and colocalization assays confirmed the interactions of DENV NS1 and human NIMA-related kinase 2 (NEK2), thousand and one amino acid protein kinase 1 (TAO1), and component of oligomeric Golgi complex 1 (COG1) proteins in virus-infected cells. This study reports a novel set of DENV NS1-interacting host cell proteins in the HepG2 cell line and proposes possible roles for human NEK2, TAO1, and COG1 in DENV infection.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Dengue Virus/metabolism , Host-Pathogen Interactions , NIMA-Related Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Dengue Virus/genetics , Gene Expression Regulation , Hep G2 Cells , Humans , NIMA-Related Kinases/genetics , Protein Binding , Protein Interaction Mapping , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Tandem Mass Spectrometry , Viral Nonstructural Proteins/genetics , Virus Replication
19.
PLoS One ; 11(2): e0149486, 2016.
Article in English | MEDLINE | ID: mdl-26901653

ABSTRACT

Dengue virus (DENV) infection causes organ injuries, and the liver is one of the most important sites of DENV infection, where viral replication generates a high viral load. The molecular mechanism of DENV-induced liver injury is still under investigation. The mitogen activated protein kinases (MAPKs), including p38 MAPK, have roles in the hepatic cell apoptosis induced by DENV. However, the in vivo role of p38 MAPK in DENV-induced liver injury is not fully understood. In this study, we investigated the role of SB203580, a p38 MAPK inhibitor, in a mouse model of DENV infection. Both the hematological parameters, leucopenia and thrombocytopenia, were improved by SB203580 treatment and liver transaminases and histopathology were also improved. We used a real-time PCR microarray to profile the expression of apoptosis-related genes. Tumor necrosis factor α, caspase 9, caspase 8, and caspase 3 proteins were significantly lower in the SB203580-treated DENV-infected mice than that in the infected control mice. Increased expressions of cytokines including TNF-α, IL-6 and IL-10, and chemokines including RANTES and IP-10 in DENV infection were reduced by SB203580 treatment. DENV infection induced the phosphorylation of p38MAPK, and its downstream signals including MAPKAPK2, HSP27 and ATF-2. SB203580 treatment did not decrease the phosphorylation of p38 MAPK, but it significantly reduced the phosphorylation of MAPKAPK2, HSP27, and ATF2. Therefore, SB203580 modulates the downstream signals to p38 MAPK and reduces DENV-induced liver injury.


Subject(s)
Activating Transcription Factor 2/metabolism , Dengue Virus/pathogenicity , HSP27 Heat-Shock Proteins/metabolism , Imidazoles/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism , Liver Diseases/drug therapy , MAP Kinase Signaling System/drug effects , Protein Serine-Threonine Kinases/metabolism , Pyridines/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Liver/drug effects , Liver/injuries , Liver/virology , Liver Diseases/virology , Male , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects
20.
PLoS One ; 10(6): e0130065, 2015.
Article in English | MEDLINE | ID: mdl-26090672

ABSTRACT

Rearrangement of membrane structure induced by dengue virus (DENV) is essential for replication, and requires host cellular machinery. Adaptor protein complex (AP)-1 is a host component, which can be recruited to components required for membrane rearrangement. Therefore, dysfunction of AP-1 may affect membrane organization, thereby decreasing replication of virus in infected cells. In the present study, AP-1-dependent traffic inhibitor inhibited DENV protein expression and virion production. We further clarified the role of AP-1A in the life cycle of DENV by RNA interference. AP-1A was not involved in DENV entry into cells. However, it facilitated DENV RNA replication. Viral RNA level was reduced significantly in Huh7 cells transfected with AP-1A small interfering RNA (siRNA) compared with control siRNA. Transfection of naked DENV viral RNA into Huh7 cells transfected with AP-1A siRNA resulted in less viral RNA and virion production than transfection into Huh7 cells transfected with control siRNA. Huh7 cells transfected with AP-1A siRNA showed greater modification of membrane structures and fewer vesicular packets compared with cells transfected with control siRNA. Therefore, AP-1A may partly control DENV-induced rearrangement of membrane structures required for viral replication.


Subject(s)
Adaptor Protein Complex 1/metabolism , Dengue Virus/physiology , Virus Replication , Adaptor Protein Complex 1/genetics , Cell Line , Dengue/metabolism , Dengue/virology , Dengue Virus/classification , Gene Knockdown Techniques , Gene Silencing , Genome, Viral , Humans , Protein Binding , Protein Transport , RNA Transport , RNA, Viral/biosynthesis , RNA, Viral/genetics , Virion/physiology , Virus Attachment , Virus Internalization , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...