Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
PLoS Pathog ; 19(9): e1011620, 2023 09.
Article in English | MEDLINE | ID: mdl-37656756

ABSTRACT

LGP2 is a RIG-I-like receptor (RLR) known to bind and recognize the intermediate double-stranded RNA (dsRNA) during virus infection and to induce type-I interferon (IFN)-related antiviral innate immune responses. Here, we find that LGP2 inhibits Zika virus (ZIKV) and tick-borne encephalitis virus (TBEV) replication independent of IFN induction. Co-immunoprecipitation (Co-IP) and confocal immunofluorescence data suggest that LGP2 likely colocalizes with the replication complex (RC) of ZIKV by interacting with viral RNA-dependent RNA polymerase (RdRP) NS5. We further verify that the regulatory domain (RD) of LGP2 directly interacts with RdRP of NS5 by biolayer interferometry assay. Data from in vitro RdRP assays indicate that LGP2 may inhibit polymerase activities of NS5 at pre-elongation but not elongation stages, while an RNA-binding-defective LGP2 mutant can still inhibit RdRP activities and virus replication. Taken together, our work suggests that LGP2 can inhibit flavivirus replication through direct interaction with NS5 protein and downregulates its polymerase pre-elongation activities, demonstrating a distinct role of LGP2 beyond its function in innate immune responses.


Subject(s)
Encephalitis Viruses, Tick-Borne , Zika Virus Infection , Zika Virus , Humans , RNA-Dependent RNA Polymerase/genetics , Nucleotidyltransferases , RNA, Double-Stranded
2.
J Gen Virol ; 105(1)2024 01.
Article in English | MEDLINE | ID: mdl-38180473

ABSTRACT

Nipah virus (NiV) is a deadly zoonotic pathogen with high potential to cause another pandemic. Owing to biosafety concerns, studies on living NiV must be performed in biosafety level 4 (BSL-4) laboratories, which greatly hinders the development of anti-NiV drugs. To overcome this issue, minigenome systems have been developed to study viral replication and screen for antiviral drugs. This study aimed to develop two minigenome systems (transient and stable expression) based on a helper cell line expressing the NiV P, N and L proteins required to initiate NiV RNA replication. Stable minigenome cells were resistant to ribavirin, remdesivir and favipiravir but sensitive to interferons. Cells of the transient replication system were sensitive to ribavirin and favipiravir and suitable for drug screening. Our study demonstrates a feasible and effective platform for studying NiV replication and shows great potential for high-throughput drug screening in a BSL-2 laboratory environment.


Subject(s)
Nipah Virus , Nipah Virus/genetics , Ribavirin , Virus Replication , Antiviral Agents/pharmacology
3.
J Virol ; 96(12): e0049422, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35604143

ABSTRACT

G protein subunit ß1 (GNB1), the beta subunit of the G protein family, plays an important role in regulating transmembrane signal transduction. Although a recent study has demonstrated that GNB1 can bind the matrix protein 1 (M1) to facilitate M1 transport to budding sites and promote the release of progeny influenza A virus (IAV), whether the GNB1 protein has other functions in IAV replication requires further study. Here, we found that GNB1 promoted IAV replication, as virus yield decreased in GNB1 knockdown or knockout cells. GNB1 interacted with polymerase subunits PB2, PB1, and PA. Overexpressed GNB1 facilitated PB2 binding to importin α3, α5, and α7 promoting the nuclear import of PB2, enhancing viral RNA synthesis and polymerase activity. Altogether, our results demonstrated that GNB1 positively regulates virus replication by interacting with polymerase subunits and facilitating the nuclear import of PB2, which provide novel insights into the molecular mechanism of IAV. IMPORTANCE Until now, there has been only one article on the role of GNB1 in IAV budding. No study has investigated the role of GNB1 in IAV replication. In this study, our research demonstrated that GNB1 could increase the interaction between PB2 and the importin α isoform and mediate the nuclear import of PB2. Therefore, GNB1 could promote viral replication and transcription. Our results provide a better understanding of the molecular mechanisms of viral replication and provide potential antiviral drug targets.


Subject(s)
Active Transport, Cell Nucleus , GTP-Binding Protein beta Subunits , Influenza A virus , Influenza, Human , Viral Proteins , GTP-Binding Protein beta Subunits/metabolism , Humans , Influenza A virus/genetics , Influenza A virus/physiology , Influenza, Human/genetics , Karyopherins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
4.
J Med Virol ; 95(3): e28657, 2023 03.
Article in English | MEDLINE | ID: mdl-36912367

ABSTRACT

Novel immune escape variants have emerged as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread worldwide. Many of the variants cause breakthrough infections in vaccinated populations, posing great challenges to current antiviral strategies targeting the immunodominance of the receptor-binding domain within the spike protein. Here, we found that a novel broadly neutralizing monoclonal antibody (mAb), G5, provided efficient protection against SARS-CoV-2 variants of concern (VOCs) in vitro and in vivo. A single dose of mAb G5 could significantly inhibit the viral burden in mice challenged with the mouse-adapted SARS-CoV-2 or SARS-CoV-2 Omicron BA.1 variant, as well as the body weight loss and cytokine release induced by mouse-adapted SARS-CoV-2. The refined epitope recognized by mAb G5 was identified as 1148 FKEELDKYF1156 in the stem helix of subunit S2. In addition, a human-mouse chimeric mAb was generated based on the variable region of heavy chain and VL genes of mAb G5. Our study provides a broad antibody drug candidate against SARS-CoV-2 VOCs and reveals a novel target for developing pan-SARS-CoV-2 vaccines.


Subject(s)
Antibodies, Monoclonal , COVID-19 , Humans , Animals , Mice , Antibodies, Monoclonal/therapeutic use , COVID-19 Vaccines , SARS-CoV-2/genetics , Immunosuppressive Agents , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing , Antibodies, Viral/therapeutic use
5.
Clin Infect Dis ; 73(3): 361-366, 2021 08 02.
Article in English | MEDLINE | ID: mdl-32638022

ABSTRACT

BACKGROUND: Currently, there is no direct evidence to prove the active replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the intestinal tract and relevant pathological changes in the colon and rectum. We investigated the presence of virions and pathological changes in surgical rectal tissues of a patient with clinically confirmed coronavirus disease 2019 (COVID-19) with rectal adenocarcinoma. METHODS: The clinical data were collected during hospitalization and follow-up of this patient. Quantitative reverse transcriptase-polymerasechain reaction (RT-PCR) was performed on the rectal tissue specimens obtained from surgical resection, succus entericus and intestinal mucosa of ileostomy, and rectal mucosa during follow-up after recovery. Ultrathin sections of surgical samples were observed for SARS-CoV-2 virions using electron microscopy. Histopathological examination was performed using hematoxylin-eosin stain. Immunohistochemical analysis and immunofluorescence were carried out on rectal tissues to evaluate the distribution of SARS-CoV-2 antigen and immune cell infiltrations. RESULTS: The patient had fever and cough on day 3 postoperatively, was diagnosed with COVID-19 on day 7, and was discharged from the hospital on day 41. RNA of SARS-CoV-2 was detected in surgically resected rectal specimens but not in samples collected 37 days after discharge. Notably, coincident with rectal tissues of surgical specimens testing nucleic acid positive for SARS-CoV-2, typical coronavirus virions in rectal tissue were observed under electron microscopy. Moreover, abundant lymphocytes and macrophages (some were SARS-CoV-2 positive) infiltrating the lamina propria were found with no significant mucosal damage. CONCLUSIONS: We first report the direct evidence of active SARS-CoV-2 replication in a patient's rectum during the incubation period, which might explain SARS-CoV-2 fecal-oral transmission.


Subject(s)
COVID-19 , SARS-CoV-2 , Hospitalization , Humans , Intestines , Patient Discharge , RNA, Viral
6.
J Nanobiotechnology ; 19(1): 295, 2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34583708

ABSTRACT

Fluorescent labeling and dynamic tracking is a powerful tool for exploring virus infection mechanisms. However, for small-sized viruses, virus tracking studies are usually hindered by a lack of appropriate labeling methods that do not dampen virus yield or infectivity. Here, we report a universal strategy for labeling viruses with chemical dyes and Quantum dots (QDs). Enterovirus 71 (EV71) was produced in a cell line that stably expresses a mutant methionyl-tRNA synthetase (MetRS), which can charge azidonorleucine (ANL) to the methionine sites of viral proteins during translation. Then, the ANL-containing virus was easily labeled with DBCO-AF647 and DBCO-QDs. The labeled virus shows sufficient yield and no obvious decrease in infectivity and can be used for imaging the virus entry process. Using the labeled EV71, different functions of scavenger receptor class B, member 2 (SCARB2), and heparan sulfate (HS) in EV71 infection were comparatively studied. The cell entry process of a strong HS-binding EV71 strain was investigated by real-time dynamic visualization of EV71-QDs in living cells. Taken together, our study described a universal biocompatible virus labeling method, visualized the dynamic viral entry process, and reported details of the receptor usage of EV71.


Subject(s)
Enterovirus/metabolism , Quantum Dots/chemistry , Receptors, Virus/metabolism , Animals , Azides , Cell Line , Chlorocebus aethiops , Enterovirus/genetics , Enterovirus A, Human/genetics , Enterovirus A, Human/metabolism , HeLa Cells , Humans , Norleucine/analogs & derivatives , Receptors, Scavenger/metabolism , Vero Cells , Viral Proteins , Virus Internalization
7.
Anal Chem ; 91(23): 15099-15106, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31698906

ABSTRACT

Detection of viruses with high sensitivity is critical for the prevention and treatment of the related disease. Two homogeneous target-induced cascade amplification methods were proposed for the detection of enterovirus 71 and coxsackievirus B3. These methods both employ DNAzyme but differ in the way in which the DNAzyme is amplified. In the hybridization chain reaction (HCR)-based strategy, the DNAzyme is assembled by hairpin DNA strands, while in the rolling circle amplification (RCA)-based strategy, the DNAzyme is synthesized by the polymerase. On the basis of the virion structure, we investigated the effects of using only VP1-antibody or VP1-antibody and VP2-antibody on the detection. And the combination of two kinds of antibodies was found to further improve the performance of the detection. Subsequently, the simultaneous detection of EV71 and CVB3 was achieved by the RCA-based strategy. And the proposed methods were also applied in clinical samples analysis with a satisfactory result, showing great potential for applications in virus detection.


Subject(s)
DNA, Catalytic/biosynthesis , Enterovirus A, Human/isolation & purification , Enterovirus B, Human/isolation & purification , Nucleic Acid Amplification Techniques/methods , Antibodies, Viral , DNA, Catalytic/metabolism , Humans
9.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29925661

ABSTRACT

Zika virus (ZIKV) infection during the large epidemics in the Americas is related to congenital abnormities or fetal demise. To date, there is no vaccine, antiviral drug, or other modality available to prevent or treat Zika virus infection. Here we designed novel live attenuated ZIKV vaccine candidates using a codon pair deoptimization strategy. Three codon pair-deoptimized ZIKVs (Min E, Min NS1, and Min E+NS1) were de novo synthesized and recovered by reverse genetics and contained large amounts of underrepresented codon pairs in the E gene and/or NS1 gene. The amino acid sequence was 100% unchanged. The codon pair-deoptimized variants had decreased replication fitness in Vero cells (Min NS1 ≫ Min E > Min E+NS1), replicated more efficiently in insect cells than in mammalian cells, and demonstrated diminished virulence in a mouse model. In particular, Min E+NS1, the most restrictive variant, induced sterilizing immunity with a robust neutralizing antibody titer, and a single immunization achieved complete protection against lethal challenge and vertical ZIKV transmission during pregnancy. More importantly, due to the numerous synonymous substitutions in the codon pair-deoptimized strains, reversion to wild-type virulence through gradual nucleotide sequence mutations is unlikely. Our results collectively demonstrate that ZIKV can be effectively attenuated by codon pair deoptimization, highlighting the potential of Min E+NS1 as a safe vaccine candidate to prevent ZIKV infections.IMPORTANCE Due to unprecedented epidemics of Zika virus (ZIKV) across the Americas and the unexpected clinical symptoms, including Guillain-Barré syndrome, microcephaly, and other birth defects in humans, there is an urgent need for ZIKV vaccine development. Here we provided the first attenuated versions of ZIKV with two important genes (E and/or NS1) that were subjected to codon pair deoptimization. Compared to parental ZIKV, the codon pair-deoptimized ZIKVs were mammal attenuated and preferred insect to mammalian cells. Min E+NS1, the most restrictive variant, induced sterilizing immunity with a robust neutralizing antibody titer and achieved complete protection against lethal challenge and vertical virus transmission during pregnancy. More importantly, the massive synonymous mutational approach made it impossible for the variant to revert to wild-type virulence. Our results have proven the feasibility of codon pair deoptimization as a strategy to develop live attenuated vaccine candidates against flaviviruses such as ZIKV, Japanese encephalitis virus, and West Nile virus.


Subject(s)
Codon/genetics , Viral Vaccines/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chlorocebus aethiops , Disease Models, Animal , Humans , Infectious Disease Transmission, Vertical/prevention & control , Mice , Reverse Genetics/methods , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vero Cells , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virulence/genetics , Virus Replication/genetics , Virus Replication/immunology , Zika Virus/genetics , Zika Virus/pathogenicity , Zika Virus Infection/immunology
10.
J Gen Virol ; 99(1): 73-85, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29182509

ABSTRACT

Enterovirus A71 (EV-A71) is a positive-strand RNA virus that causes hand-foot-mouth disease and neurological complications in children and infants. Although the underlying mechanisms remain to be further defined, impaired immunity is thought to play an important role. The host zinc-finger antiviral protein (ZAP), an IFN-stimulated gene product, has been reported to specifically inhibit the replication of certain viruses. However, whether ZAP restricts the infection of enteroviruses remains unknown. Here, we report that EV-A71 infection upregulates ZAP mRNA in RD and HeLa cells. Moreover, ZAP overexpression rendered 293 T cells resistant to EV-A71 infection, whereas siRNA-mediated depletion of endogenous ZAP enhanced EV-A71 infection. The EV-A71 infection stimulated site-specific proteolysis of two ZAP isoforms, leading to the accumulation of a 40 kDa N-terminal ZAP fragment in virus-infected cells. We further revealed that the 3C protease (3Cpro) of EV-A71 mediates ZAP cleavage, which requires protease activity. Furthermore, ZAP variants with single amino acid substitutions at Gln-369 were resistant to 3Cpro cleavage, implying that Gln-369 is the sole cleavage site in ZAP. Moreover, although ZAP overexpression inhibited EV-A71 replication, the cleaved fragments did not show this effect. Our results indicate that an equilibrium between ZAP and enterovirus 3Cpro controls viral infection. The findings in this study suggest that viral 3Cpro mediated ZAP cleavage may represent a mechanism to escape host antiviral responses.


Subject(s)
Cysteine Endopeptidases/metabolism , Enterovirus A, Human/enzymology , Host-Pathogen Interactions , RNA-Binding Proteins/metabolism , Viral Proteins/metabolism , Virus Replication , 3C Viral Proteases , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Enterovirus A, Human/genetics , Gene Expression Regulation , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Muscle Cells/metabolism , Muscle Cells/virology , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Sf9 Cells/immunology , Sf9 Cells/virology , Signal Transduction , Spodoptera , Viral Proteins/genetics
11.
J Neuroinflammation ; 15(1): 275, 2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30241539

ABSTRACT

BACKGROUND: Many viruses depend on the extensive membranous network of the endoplasmic reticulum (ER) for their translation, replication, and packaging. Certain membrane modifications of the ER can be a trigger for ER stress, as well as the accumulation of viral protein in the ER by viral infection. Then, unfolded protein response (UPR) is activated to alleviate the stress. Zika virus (ZIKV) is a mosquito-borne flavivirus and its infection causes microcephaly in newborns and serious neurological complications in adults. Here, we investigated ER stress and the regulating model of UPR in ZIKV-infected neural cells in vitro and in vivo. METHODS: Mice deficient in type I and II IFN receptors were infected with ZIKV via intraperitoneal injection and the nervous tissues of the mice were assayed at 5 days post-infection. The expression of phospho-IRE1, XBP1, and ATF6 which were the key markers of ER stress were analyzed by immunohistochemistry assay in vivo. Additionally, the nuclear localization of XBP1s and ATF6n were analyzed by immunohistofluorescence. Furthermore, two representative neural cells, neuroblastoma cell line (SK-N-SH) and astrocytoma cell line (CCF-STTG1), were selected to verify the ER stress in vitro. The expression of BIP, phospho-elF2α, phospho-IRE1, and ATF6 were analyzed through western blot and the nuclear localization of XBP1s was performed by confocal immunofluorescence microscopy. RT-qPCR was also used to quantify the mRNA level of the UPR downstream genes in vitro and in vivo. RESULTS: ZIKV infection significantly upregulated the expression of ER stress markers in vitro and in vivo. Phospho-IRE1 and XBP1 expression significantly increased in the cerebellum and mesocephalon, while ATF6 expression significantly increased in the mesocephalon. ATF6n and XBP1s were translocated into the cell nucleus. The levels of BIP, ATF6, phospho-elf2α, and spliced xbp1 also significantly increased in vitro. Furthermore, the downstream genes of UPR were detected to investigate the regulating model of the UPR during ZIKV infection in vitro and in vivo. The transcriptional levels of atf4, gadd34, chop, and edem-1 in vivo and that of gadd34 and chop in vitro significantly increased. CONCLUSION: Findings in this study demonstrated that ZIKV infection activates ER stress in neural cells. The results offer clues to further study the mechanism of neuropathogenesis caused by ZIKV infection.


Subject(s)
Activating Transcription Factor 6/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , X-Box Binding Protein 1/metabolism , Zika Virus Infection/metabolism , Zika Virus Infection/pathology , Animals , Brain/metabolism , Brain/pathology , Brain/virology , Cell Line, Transformed , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/virology , Protein Serine-Threonine Kinases/genetics , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , X-Box Binding Protein 1/genetics , Zika Virus/genetics , Zika Virus/physiology
12.
J Virol ; 90(10): 5141-5151, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962213

ABSTRACT

UNLABELLED: The NF-κB signaling network, which is an ancient signaling pathway, plays a pivotal role in innate immunity and constitutes a first line of defense against invading pathogens, including viruses. However, numerous viruses possess evolved strategies to antagonize the activation of the NF-κB signaling pathway. Our previous study demonstrated that the nonstructural protein 2C of enterovirus 71 (EV71), which is the major pathogen of hand, foot, and mouth disease, inhibits tumor necrosis factor alpha (TNF-α)-mediated activation of NF-κB by suppressing IκB kinase ß (IKKß) phosphorylation. Nevertheless, the mechanism underlying the inhibition of IKKß phosphorylation by EV71 2C remains largely elusive. We demonstrate that EV71 2C interacts with all isoforms of the protein phosphatase 1 (PP1) catalytic subunit (the PP1α, PP1ß, and PP1γ isoforms) through PP1-docking motifs. EV71 2C has no influence on the subcellular localization of PP1. In addition, the PP1-binding-deficient EV71 2C mutant 3E3L nearly completely lost the ability to suppress IKKß phosphorylation and NF-κB activation was markedly restored in the mutant, thereby indicating that PP1 binding is efficient for EV71 2C-mediated inhibition of IKKß phosphorylation and NF-κB activation. We further demonstrate that 2C forms a complex with PP1 and IKKß to dephosphorylate IKKß. Notably, we reveal that other human enteroviruses, including poliovirus (PV), coxsackie A virus 16 (CVA16), and coxsackie B virus 3 (CVB3), use 2C proteins to recruit PP1, leading to the inhibition of IKKß phosphorylation. Our findings indicate that enteroviruses exploit a novel mechanism to inhibit IKKß phosphorylation by recruiting PP1 and IKKß to form a complex through 2C proteins, which ultimately results in the inhibition of the NF-κB signaling pathway. IMPORTANCE: The innate antiviral immunity system performs an essential function in recognizing and eliminating invading viruses. Enteroviruses include a number of important human pathogens, including poliovirus (PV), EV71, and coxsackieviruses (CVs). As 2C is the most conserved and complex nonstructural protein of enteroviruses, its biological function is largely unclear, whereas the 2A and 3C proteinases of enteroviruses are well characterized. We reveal that EV71 2C forms a complex with PP1 and IKKß to maintain IKKß in an unphosphorylated and inactive state, resulting in the inactivation of the TNF-α-mediated NF-κB signaling pathway. We provide evidence that the 2C proteins of the enteroviruses PV, CVA16, and CVB3 suppress IKKß phosphorylation through the same mechanism involving PP1. We demonstrate that enteroviruses exploit a novel mechanism involving PP1 to regulate innate antiviral immunity, and our findings may be particularly important for understanding the pathogenicity of enteroviruses.


Subject(s)
Carrier Proteins/metabolism , Enterovirus/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Protein Phosphatase 1/metabolism , Viral Nonstructural Proteins/metabolism , Carrier Proteins/genetics , Enterovirus/chemistry , Enterovirus/genetics , Enterovirus A, Human/chemistry , Enterovirus A, Human/metabolism , Enterovirus B, Human/metabolism , HeLa Cells , Humans , Phosphorylation , Poliovirus/chemistry , Poliovirus/metabolism , Protein Binding , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Viral Nonstructural Proteins/genetics
13.
J Virol ; 90(23): 10472-10485, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27630238

ABSTRACT

Accumulating evidence suggests that viruses hijack cellular proteins to circumvent the host immune system. Ubiquitination and SUMOylation are extensively studied posttranslational modifications (PTMs) that play critical roles in diverse biological processes. Cross talk between ubiquitination and SUMOylation of both host and viral proteins has been reported to result in distinct functional consequences. Enterovirus 71 (EV71), an RNA virus belonging to the family Picornaviridae, is a common cause of hand, foot, and mouth disease. Little is known concerning how host PTM systems interact with enteroviruses. Here, we demonstrate that the 3D protein, an RNA-dependent RNA polymerase (RdRp) of EV71, is modified by small ubiquitin-like modifier 1 (SUMO-1) both during infection and in vitro Residues K159 and L150/D151/L152 were responsible for 3D SUMOylation as determined by bioinformatics prediction combined with site-directed mutagenesis. Also, primer-dependent polymerase assays indicated that mutation of SUMOylation sites impaired 3D polymerase activity and virus replication. Moreover, 3D is ubiquitinated in a SUMO-dependent manner, and SUMOylation is crucial for 3D stability, which may be due to the interplay between the two PTMs. Importantly, increasing the level of SUMO-1 in EV71-infected cells augmented the SUMOylation and ubiquitination levels of 3D, leading to enhanced replication of EV71. These results together suggested that SUMO and ubiquitin cooperatively regulated EV71 infection, either by SUMO-ubiquitin hybrid chains or by ubiquitin conjugating to the exposed lysine residue through SUMOylation. Our study provides new insight into how a virus utilizes cellular pathways to facilitate its replication. IMPORTANCE: Infection with enterovirus 71 (EV71) often causes neurological diseases in children, and EV71 is responsible for the majority of fatalities. Based on a better understanding of interplay between virus and host cell, antiviral drugs against enteroviruses may be developed. As a dynamic cellular process of posttranslational modification, SUMOylation regulates global cellular protein localization, interaction, stability, and enzymatic activity. However, little is known concerning how SUMOylation directly influences virus replication by targeting viral polymerase. Here, we found that EV71 polymerase 3D was SUMOylated during EV71 infection and in vitro Moreover, the SUMOylation sites were determined, and in vitro polymerase assays indicated that mutations at SUMOylation sites could impair polymerase synthesis. Importantly, 3D is ubiquitinated in a SUMOylation-dependent manner that enhances the stability of the viral polymerase. Our findings indicate that the two modifications likely cooperatively enhance virus replication. Our study may offer a new therapeutic strategy against virus replication.


Subject(s)
Enterovirus A, Human/physiology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Chlorocebus aethiops , Enterovirus A, Human/genetics , Enzyme Stability , HEK293 Cells , Host-Pathogen Interactions , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sumoylation , Ubiquitination , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/physiology
14.
Analyst ; 142(4): 608-612, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28138659

ABSTRACT

Herein a convenient strategy was developed to quantify protein SUMOylation. The amount of target proteins and SUMO proteins conjugated to them could be quantified through luciferase-based bioluminescence detection and the relative bioluminescence was used to evaluate the SUMOylation degree of the target proteins.


Subject(s)
Biosensing Techniques , Luciferases/chemistry , Small Ubiquitin-Related Modifier Proteins/chemistry , Sumoylation , HEK293 Cells , Humans
15.
Biochem Biophys Res Commun ; 467(4): 760-5, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26482854

ABSTRACT

Baculoviruses are potential vectors of gene therapy for the ability to transfer gene high efficiently into mammalian cells. However, cell membrane proteins which interact with baculoviral glycoproteins have not been identified. In this study, we developed a self-biotinylated AcMNPV bearing biotinylated GP64 glycoproteins. This recombinant virus demonstrated the capability to infect insect cells and to transduct mammalian cells. Using this biotinylated virus, a protein >170Kda which could specifically interact with GP64 proteins was identified from virus transducted BHK-21 cells through cross-linking and streptavidin purification. Our study provides a useful approach for identifying cell membrane proteins that interact with baculovirus surface proteins or proteins involved in virus attachment.


Subject(s)
Membrane Glycoproteins/metabolism , Nucleopolyhedroviruses/genetics , Proteins/metabolism , Animals , Biotinylation , Cell Line , Cricetinae , Cross-Linking Reagents/chemistry , Genetic Engineering/methods , Genetic Vectors , Kidney/cytology , Kidney/virology , Membrane Glycoproteins/genetics , Nucleopolyhedroviruses/metabolism , Proteins/analysis , Succinimides/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
16.
J Immunol ; 191(2): 660-9, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772026

ABSTRACT

Precise regulation of innate immunity is crucial for maintaining optimal immune responses against infections. Whereas positive regulation of IFN signaling elicits rapid type I IFNs, negative regulation is equally important in preventing the production of superfluous IFNs that can be hazardous to the host. The positive regulators of IFN pathway are known to be the main targets of viruses to antagonize the innate immune system. Whether viruses target the negative regulators of IFN pathway remains to be fully investigated. In this study, we report that the structural protein VP2 of human Bocavirus modulates IFN pathway by targeting the ring finger protein 125 (RNF125), a negative regulator of type I IFN signaling, which conjugates Lys(48)-linked ubiquitination to retinoic acid-inducible gene-I (RIG-I) and subsequently leads to the proteasome-dependent degradation of RIG-I. VP2 not only upregulated Sendai virus (SeV)-induced IFNB promoter activity, but also enhanced SeV-induced IFN-ß production at both mRNA and protein levels. In agreement, the level of Ser(396)-phosphorylated IFN regulatory factor 3 stimulated by SeV was enhanced in the presence of VP2. Furthermore, VP2 was demonstrated to physically interact with RNF125, resulting in the reduction of RNF125-mediated ubiquitination and proteasome-dependent degradation of RIG-I. Additional study indicated that endogenous RIG-I degradation was decreased in VP2-expressing cells. Our study delineates a unique phenomenon for aberrant activation of IFN regulatory factor 3 pathway and may represent a new mechanism underlying viral manipulation of the host immune system.


Subject(s)
Capsid Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Human bocavirus , Interferon-beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , DEAD Box Protein 58 , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Immunologic , Sendai virus , Signal Transduction , Ubiquitination
17.
J Virol ; 87(10): 5645-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23468506

ABSTRACT

Enterovirus 71 (EV71) has emerged as a major cause of neurological disease following the near eradication of poliovirus. Accumulating evidence suggests that mammalian microRNAs (miRNAs), a class of noncoding RNAs of 18 to 23 nucleotides (nt) with important regulatory roles in many cellular processes, participate in host antiviral defenses. However, the roles of miRNAs in EV71 infection and pathogenesis are still unclear. Here, hsa-miR-296-5p expression was significantly increased in EV71-infected human cells. As determined by virus titration, quantitative real-time PCR (qRT-PCR), and Western blotting, overexpression of hsa-miR-296-5p inhibited, while inhibition of endogenous hsa-miR-296-5p facilitated, EV71 infection. Additionally, two potential hsa-miR-296-5p targets (nt 2115 to 2135 and nt 2896 to 2920) located in the EV71 genome (strain BrCr) were bioinformatically predicted and validated by luciferase reporter assays and Western blotting. Genomic alignment of various EV71 strains revealed synonymous mutations in hsa-miR-296-5p target sequences. Furthermore, the introduction of synonymous mutations into the EV71 BrCr genome by site-directed mutagenesis impaired the viral inhibitory effects of hsa-miR-296-5p and facilitated mutant virus infection. Meanwhile, compensatory mutations in corresponding hsa-miR-296-5p target sequences of the EV71 HeN strain (GenBank accession number JN256064) restored the inhibitory effects of the miRNA. These results indicate that hsa-miR-296-5p inhibits EV71 replication by targeting the viral genome. Our findings support the notion that cellular miRNAs can inhibit virus infection and that the virus mutates to escape suppression by cellular miRNAs.


Subject(s)
Enterovirus A, Human/immunology , Enterovirus A, Human/physiology , Host-Pathogen Interactions , MicroRNAs/metabolism , Virus Replication , Animals , Binding Sites , Blotting, Western , Cell Line , Gene Expression , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Up-Regulation , Viral Load
18.
J Immunol ; 189(3): 1144-53, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22745372

ABSTRACT

Human bocavirus (HBoV) mainly infects young children. Although many infected children suffer from respiratory or gastroenteric tract diseases, an association between HBoV and these diseases is not definite. Because modulation of type I IFN is crucial for viruses to establish efficient replication, in this study, we tested whether HBoV modulates type I IFN production. We observed that a nearly full-length HBoV clone significantly reduced both Sendai virus (SeV)- and poly(deoxyadenylic-thymidylic) acid-induced IFN-ß production. Further study showed that NP1 blocked IFN-ß activation in response to SeV, poly(deoxyadenylic-thymidylic) acid, and IFN-ß pathway inducers, including retinoic acid-inducible protein I, mitochondrial antiviral signaling protein, inhibitor of κB kinase ε, and TANK-binding kinase 1. In addition, NP1 interfered with IRF-3-responsive PRD(III-I) promoter activated by SeV and a constitutively active mutant of IRF-3 (IRF-3/5D). Although NP1 suppressed the IRF-3 pathway, it did not affect IRF-3 activation processes, including phosphorylation, dimerization, and nuclear translocation. Coimmunoprecipitation assays confirmed the interaction between NP1 and IRF-3. Additional deletion mutagenesis and coimmunoprecipitation assays revealed that NP1 bound to the DNA-binding domain of IRF-3, resulting in the interruption of an association between IRF-3 and IFNB promoter. Altogether, our results indicate that HBoV NP1 blocks IFN production through a unique mechanism. To our knowledge, this is the first study to investigate the modulation of innate immunity by HBoV. Our findings suggest a potential immune-evasion mechanism used by HBoV and provide a basis for better understanding HBoV pathogenesis.


Subject(s)
Bocavirus/immunology , Host-Pathogen Interactions , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/metabolism , Interferon-beta/antagonists & inhibitors , Interferon-beta/biosynthesis , Promoter Regions, Genetic/immunology , Viral Nonstructural Proteins/physiology , Cell Line, Tumor , HEK293 Cells , Humans , Interferon-beta/genetics , Signal Transduction/immunology
19.
Virol Sin ; 39(1): 56-70, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37967718

ABSTRACT

Avian H9N2 viruses have wide host range among the influenza A viruses. However, knowledge of H9N2 mammalian adaptation is limited. To explore the molecular basis of the adaptation to mammals, we performed serial lung passaging of the H9N2 strain A/chicken/Hunan/8.27 YYGK3W3-OC/2018 (3W3) in mice and identified six mutations in the hemagglutinin (HA) and polymerase acidic (PA) proteins. Mutations L226Q, T511I, and A528V of HA were responsible for enhanced pathogenicity and viral replication in mice; notably, HA-L226Q was the key determinant. Mutations T97I, I545V, and S594G of PA contributed to enhanced polymerase activity in mammalian cells and increased viral replication levels in vitro and in vivo. PA-T97I increased viral polymerase activity by accelerating the viral polymerase complex assembly. Our findings revealed that the viral replication was affected by the presence of PA-97I and/or PA-545V in combination with a triple-point HA mutation. Furthermore, the double- and triple-point PA mutations demonstrated antagonistic effect on viral replication when combined with HA-226Q. Notably, any combination of PA mutations, along with double-point HA mutations, resulted in antagonistic effect on viral replication. We also observed antagonism in viral replication between PA-545V and PA-97I, as well as between HA-528V and PA-545V. Our findings demonstrated that several antagonistic mutations in HA and PA proteins affect viral replication, which may contribute to the H9N2 virus adaptation to mice and mammalian cells. These findings can potentially contribute to the monitoring of H9N2 field strains for assessing their potential risk in mammals.


Subject(s)
Influenza A Virus, H9N2 Subtype , Influenza in Birds , Orthomyxoviridae Infections , Animals , Mice , Influenza A Virus, H9N2 Subtype/genetics , Hemagglutinins , Viral Proteins/genetics , Viral Proteins/metabolism , Mutation , Virus Replication/genetics , Nucleotidyltransferases , Chickens , Mammals/metabolism
20.
J Gen Virol ; 94(Pt 6): 1335-1342, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23388199

ABSTRACT

Human bocavirus (HBoV), closely related to canine minute virus (MVC) and bovine parvovirus (BPV), is a new member of the Bocavirus genus within the Parvoviridae family. The non-structural protein NP1 of HBoV is a nuclear localized protein and plays an important role in DNA replication as well as in the evasion of host innate immunity. In the current study, we provide the first evidence that NP1 possesses a non-classical nuclear localization signal (ncNLS) (amino acids 7-50). Embedded within this ncNLS is a classical bipartite nuclear localization signal (cNLS) (amino acids 14-28), capable of transporting a heterologous cytoplasmic protein ß-galactosidase fusion protein (ß-gal-EGFP) to the nucleus via the classical importin α/ß1-mediated pathway. Amino acids 7-50 containing the cNLS and the ncNLS of NP1 or full-length NP1 interact with importin α1, importin ß1 and importin ß1Δ, which lacks the importin α binding domain, indicating that the nuclear import of NP1 is through both conventional importin α/ß1 heterodimer- and non-classical importinß1-mediated pathways. Given that the arrangement of a cNLS embedded within an ncNLS is unusual in viral proteins, our data together reveal a novel molecular mechanism underlying the nuclear import of HBoV NP1, providing a basis for further understanding its biological function.


Subject(s)
Cell Nucleus/metabolism , Human bocavirus/genetics , Nuclear Localization Signals , Parvoviridae Infections/virology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Active Transport, Cell Nucleus , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Cell Nucleus/virology , Human bocavirus/chemistry , Human bocavirus/metabolism , Humans , Parvoviridae Infections/metabolism , Viral Nonstructural Proteins/genetics , beta Karyopherins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL