250 results on '"Chunfu Zheng"'
Search Results
102. Real-time quantitative PCR (qPCR) v1
- Author
-
Chunfu Zheng, Hui Yuan, Jia You, and Hongjuan You
- Published
- 2018
- Full Text
- View/download PDF
103. co-immunoprecipitation ?co-IP? v1
- Author
-
Chunfu Zheng, Hui Yuan, Jia You, and Hongjuan You
- Published
- 2018
- Full Text
- View/download PDF
104. Western blot (WB) v1
- Author
-
Chunfu Zheng, Hui Yuan, Jia You, and Hongjuan You
- Published
- 2018
- Full Text
- View/download PDF
105. Dual-Luciferase® Reporter (DLR) Assay v1
- Author
-
Chunfu Zheng, Hui Yuan, Jia You, and Hongjuan You
- Published
- 2018
- Full Text
- View/download PDF
106. Western blot v1
- Author
-
Hui Yuan and Chunfu Zheng
- Published
- 2018
- Full Text
- View/download PDF
107. Evasion of Cytosolic DNA-Stimulated Innate Immune Responses by Herpes Simplex Virus 1
- Author
-
Chunfu Zheng
- Subjects
0301 basic medicine ,viruses ,Immunology ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,Interferon ,Virology ,medicine ,Animals ,Humans ,Immune Evasion ,Gem ,Innate immune system ,Pattern recognition receptor ,Membrane Proteins ,Herpes Simplex ,Immunity, Innate ,Sting ,030104 developmental biology ,Herpes simplex virus ,chemistry ,Receptors, Pattern Recognition ,Insect Science ,Stimulator of interferon genes ,DNA, Viral ,Nucleic acid ,DNA ,Signal Transduction ,medicine.drug - Abstract
Recognition of virus-derived nucleic acids by host pattern recognition receptors (PRRs) is crucial for early defense against viral infections. Recent studies revealed that PRRs also include several newly identified DNA sensors, most of which could activate the downstream adaptor stimulator of interferon genes (STING) and lead to the production of host antiviral factors. Herpes simplex virus 1 (HSV-1) is extremely successful in establishing effective infections, due to its capacity to counteract host innate antiviral responses. In this Gem, I summarize the most recent findings on the molecular mechanisms utilized by HSV-1 to target different steps of the cellular DNA-sensor-mediated antiviral signal pathway.
- Published
- 2018
- Full Text
- View/download PDF
108. Herpes Simplex Virus 1 Abrogates the cGAS/STING-Mediated Cytosolic DNA-Sensing Pathway via Its Virion Host Shutoff Protein, UL41
- Author
-
Chunfu Zheng and Chenhe Su
- Subjects
0301 basic medicine ,viruses ,Immunology ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,03 medical and health sciences ,Viral Proteins ,Interferon ,Virology ,medicine ,Immune Evasion ,Gene knockdown ,Cytosolic DNA-Sensing Pathway ,Signal transducing adaptor protein ,Membrane Proteins ,Nucleotidyltransferases ,Immunity, Innate ,Cell biology ,Virus-Cell Interactions ,030104 developmental biology ,Herpes simplex virus ,Lytic cycle ,Insect Science ,Stimulator of interferon genes ,DNA, Viral ,Host-Pathogen Interactions ,medicine.drug - Abstract
Cyclic GMP-AMP synthase (cGAS) is a key DNA sensor capable of detecting microbial DNA and activating the adaptor protein stimulator of interferon genes (STING), leading to interferon (IFN) production and host antiviral responses. Cells exhibited reduced type I IFN production in response to cytosolic DNA in the absence of cGAS. Although the cGAS/STING-mediated DNA-sensing signal is crucial for host defense against many viruses, especially for DNA viruses, few viral components have been identified to specifically target this signaling pathway. Herpes simplex virus 1 (HSV-1) is a DNA virus that has evolved multiple strategies to evade host immune responses. In the present study, we found that HSV-1 tegument protein UL41 was involved in counteracting the cGAS/STING-mediated DNA-sensing pathway. Our results showed that wild-type (WT) HSV-1 infection could inhibit immunostimulatory DNA-induced activation of the IFN signaling pathway compared with the UL41-null mutant virus (R2621), and ectopic expression of UL41 decreased cGAS/STING-mediated IFN-β promoter activation and IFN-β production. Further study indicated that UL41 reduced the accumulation of cGAS to abrogate host recognition of viral DNA. In addition, stable knockdown of cGAS facilitated the replication of R2621 but not WT HSV-1. For the first time, HSV-1 UL41 was demonstrated to evade the cGAS/STING-mediated DNA-sensing pathway by degrading cGAS via its RNase activity. IMPORTANCE HSV-1 is well known for its ability to evade host antiviral responses and establish a lifelong latent infection while triggering reactivation and lytic infection under stress. Currently, whether HSV-1 evades the cytosolic DNA sensing and signaling is still poorly understood. In the present study, we found that tegument protein UL41 targeted the cGAS/STING-mediated cellular DNA-sensing pathway by selectively degrading cGAS mRNA. Knockdown of endogenous cGAS could facilitate the replication of R2621 but not WT HSV-1. Furthermore, UL41 was shown for the first time to act directly on cGAS. Findings in this study could provide new insights into the host-virus interaction and help develop new approaches against HSV-1.
- Published
- 2017
109. Herpes Simplex Virus 1 UL41 Protein Suppresses the IRE1/XBP1 Signal Pathway of the Unfolded Protein Response via Its RNase Activity
- Author
-
Chunfu Zheng, Chenhe Su, Pengchao Zhang, and Zhangtao Jiang
- Subjects
X-Box Binding Protein 1 ,0301 basic medicine ,Thapsigargin ,XBP1 ,RNA Splicing ,Immunology ,Herpesvirus 1, Human ,Protein Serine-Threonine Kinases ,Protein degradation ,Biology ,Endoplasmic-reticulum-associated protein degradation ,Microbiology ,Cell Line ,Ectopic Gene Expression ,Lactones ,Viral Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,Virology ,Endoribonucleases ,Humans ,RNA, Messenger ,Messenger RNA ,030102 biochemistry & molecular biology ,Endoplasmic reticulum ,Herpes Simplex ,Virus-Cell Interactions ,Cell biology ,Enzyme Activation ,030104 developmental biology ,chemistry ,Insect Science ,Unfolded Protein Response ,Unfolded protein response ,Sesquiterpenes ,Signal Transduction - Abstract
During viral infection, accumulation of viral proteins can cause stress in the endoplasmic reticulum (ER) and trigger the unfolded protein response (UPR) to restore ER homeostasis. The inositol-requiring enzyme 1 (IRE1)-dependent pathway is the most conserved of the three UPR signal pathways. Upon activation, IRE1 splices out an intron from the unspliced inactive form of X box binding protein 1 [XBP1(u)] mRNA and produces a transcriptionally potent spliced form [XBP1(s)]. Previous studies have reported that the IRE1/XBP1 pathway is inhibited upon herpes simplex virus 1 (HSV-1) infection; however, the underlying molecular mechanism is still elusive. Here, we uncovered a role of the HSV-1 UL41 protein in inhibiting the IRE1/XBP1 signal pathway. Ectopic expression of UL41 decreased the expression of XBP1 and blocked XBP1 splicing activation induced by the ER stress inducer thapsigargin. Wild-type (WT) HSV-1, but not the UL41-null mutant HSV-1 (R2621), decreased XBP1 mRNA induced by thapsigargin. Nevertheless, infection with both WT HSV-1 and R2621 without drug pretreatment could reduce the mRNA and protein levels of XBP1(s), and additional mechanisms might contribute to this inhibition of XBP1(s) during R2621 infection. Taking these findings together, our results reveal XBP1 as a novel target of UL41 and provide insights into the mechanism by which HSV-1 modulates the IRE1/XBP1 pathway. IMPORTANCE During viral infection, viruses hijack the host translation apparatus to produce large amounts of viral proteins, which leads to ER stress. To restore ER homeostasis, cells initiate the UPR to alleviate the effects of ER stress. The IRE1/XBP1 pathway is the most conserved UPR branch, and it activates ER-associated protein degradation (ERAD) to reduce the ER load. The IRE1/XBP1 branch is repressed during HSV-1 infection, but little is known about the underlying molecular mechanism. Our results show for the first time that UL41 suppresses the IRE1/XBP1 signal pathway by reducing the accumulation of XBP1 mRNA, and characterization of the underlying molecular mechanism provides new insight into the modulation of UPR by HSV-1.
- Published
- 2017
- Full Text
- View/download PDF
110. Herpes Simplex Virus 1 UL24 Abrogates the DNA Sensing Signal Pathway by Inhibiting NF-κB Activation
- Author
-
Haiyan Xu, Chunfu Zheng, Chenhe Su, Christopher H. Mody, and Angela Pearson
- Subjects
0301 basic medicine ,Gene Expression Regulation, Viral ,Immunology ,Active Transport, Cell Nucleus ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,Viral Proteins ,Transcription (biology) ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Protein Interaction Maps ,Promoter Regions, Genetic ,Gene ,Vero Cells ,Innate immune system ,030102 biochemistry & molecular biology ,Interleukin-6 ,Pattern recognition receptor ,Transcription Factor RelA ,NF-kappa B p50 Subunit ,NF-κB ,Interferon-beta ,Cell biology ,Virus-Cell Interactions ,030104 developmental biology ,Herpes simplex virus ,HEK293 Cells ,Viral replication ,chemistry ,Insect Science ,Stimulator of interferon genes ,Host-Pathogen Interactions ,HeLa Cells ,Protein Binding ,Signal Transduction - Abstract
Cyclic GMP-AMP synthase (cGAS) is a newly identified DNA sensor that recognizes foreign DNA, including the genome of herpes simplex virus 1 (HSV-1). Upon binding of viral DNA, cGAS produces cyclic GMP-AMP, which interacts with and activates stimulator of interferon genes (STING) to trigger the transcription of antiviral genes such as type I interferons (IFNs), and the production of inflammatory cytokines. HSV-1 UL24 is widely conserved among members of the herpesviruses family and is essential for efficient viral replication. In this study, we found that ectopically expressed UL24 could inhibit cGAS-STING-mediated promoter activation of IFN-β and interleukin-6 (IL-6), and UL24 also inhibited interferon-stimulatory DNA-mediated IFN-β and IL-6 production during HSV-1 infection. Furthermore, UL24 selectively blocked nuclear factor κB (NF-κB) but not IFN-regulatory factor 3 promoter activation. Coimmunoprecipitation analysis demonstrated that UL24 bound to the endogenous NF-κB subunits p65 and p50 in HSV-1-infected cells, and UL24 was also found to bind the Rel homology domains (RHDs) of these subunits. Furthermore, UL24 reduced the tumor necrosis factor alpha (TNF-α)-mediated nuclear translocation of p65 and p50. Finally, mutational analysis revealed that the region spanning amino acids (aa) 74 to 134 of UL24 [UL24(74–134)] is responsible for inhibiting cGAS-STING-mediated NF-κB promoter activity. For the first time, UL24 was shown to play an important role in immune evasion during HSV-1 infection. IMPORTANCE NF-κB is a critical component of the innate immune response and is strongly induced downstream of most pattern recognition receptors (PRRs), leading to the production of IFN-β as well as a number of inflammatory chemokines and interleukins. To establish persistent infection, viruses have evolved various mechanisms to counteract the host NF-κB pathway. In the present study, for the first time, HSV-1 UL24 was demonstrated to inhibit the activation of NF-κB in the DNA sensing signal pathway via binding to the RHDs of the NF-κB subunits p65 and p50 and abolishing their nuclear translocation.
- Published
- 2017
111. Additional file 1: Figure S1. of Antiviral activity of PHA767491 against human herpes simplex virus in vitro and in vivo
- Author
-
Hou, Jue, Zili Zhang, Huang, Qiang, Yan, Jun, Xiaohu Zhang, Xiaoliang Yu, Guihua Tan, Chunfu Zheng, Xu, Feng, and Sudan He
- Abstract
PHA767491 has no effect on NF-ÎşB and MAPK activation. (DOCX 526 kb)
- Published
- 2017
- Full Text
- View/download PDF
112. Crystal Structure of Herpes Simplex Virus 2 gD Bound to Nectin-1 Reveals a Conserved Mode of Receptor Recognition
- Author
-
Chunfu Zheng, Jinghua Yan, Guangwen Lu, Zhujun Chen, Jianxun Qi, Na Zhang, George F. Gao, and Yan Li
- Subjects
Protein Conformation ,Viral protein ,Herpesvirus 2, Human ,viruses ,DNA Mutational Analysis ,Nectins ,Immunology ,CHO Cells ,Plasma protein binding ,Biology ,Crystallography, X-Ray ,medicine.disease_cause ,Microbiology ,Cricetulus ,Protein structure ,Viral Envelope Proteins ,Nectin ,Virology ,medicine ,Animals ,Humans ,Surface plasmon resonance ,Tropism ,Structure and Assembly ,C-terminus ,Surface Plasmon Resonance ,Herpes simplex virus ,Amino Acid Substitution ,Biochemistry ,Insect Science ,Biophysics ,Receptors, Virus ,Cell Adhesion Molecules ,Protein Binding - Abstract
Herpes simplex virus 1 (HSV-1) and HSV-2 are among the most prevalent human pathogens. Both viruses can recognize, via the surface envelope glycoprotein D (gD), human nectin-1 as a functional receptor. Previous studies have successfully elucidated the molecular basis of the binding between HSV-1 gD and nectin-1 by cocrystallography. Despite a high sequence identity between HSV-1 and HSV-2 gDs, the atomic intermolecule details for the HSV-2-gD/nectin-1 interaction remain elusive. Here, we report the crystal structures of both the unbound and the nectin-1-bound HSV-2 gDs. The free-gD structure expectedly comprises an IgV-like core and the surface-exposed terminal extensions as observed in its HSV-1 counterpart but lacks traceable electron densities for a large portion of the terminal elements. These terminal residues were clearly traced in the complex structure as a definitive loop in the N terminus and an α-helix in the C terminus, thereby showing a conserved nectin-1-binding mode as reported for HSV-1 gD. The interface residues in nectin-1 were further mutated and tested for the gD interaction by surface plasmon resonance. The resultant binding patterns were similar for HSV-1 and HSV-2 gDs, further supporting a homologous receptor-binding basis by the two viruses for nectin-1. These data, together with a cell-based fusion assay showing a cross-inhibition of the gD/nectin-1-mediated cell-cell fusion by soluble HSV-1 and HSV-2 gDs, provided solid structural and functional evidence that HSV-1 and HSV-2 recognize nectin-1 via the same binding mode. Finally, we also demonstrated that nectin-1 I80 is an important residue involved in gD interaction. IMPORTANCE Despite intensified studies, a detailed picture of the molecular features in the HSV-2-gD/nectin-1 interaction remains unavailable. Previous work focused on HSV-1 gD, which folds into an IgV-like core with large terminal extensions and utilizes the extension elements to engage nectin-1. Here, we report the crystal structures of HSV-2 gD in both the free and the nectin-1-bound forms. The atomic intermolecule details for HSV-2-gD/nectin-1 interaction are clearly presented. The observed binding mode is identical to that reported for its HSV-1 counterpart. This structural observation was further supported by our comparative functional assays showing that nectin-1 mutations similarly affect the ligand-receptor interaction of both virus gDs. Taken together, we provide comprehensive structural and functional data demonstrating a conserved receptor-binding mode between HSV-1 and HSV-2 for nectin-1. Our results also indicate that the tropism difference between the two viruses likely arises from aspects other than the gD/nectin-1 binding features.
- Published
- 2014
- Full Text
- View/download PDF
113. Herpes Simplex Virus 1 Protein Kinase US3 Hyperphosphorylates p65/RelA and Dampens NF-κB Activation
- Author
-
Shuai Wang, Chunfu Zheng, Liwen Ni, and Kezhen Wang
- Subjects
Chemokine ,viruses ,Immunology ,Mutation, Missense ,Down-Regulation ,Herpesvirus 1, Human ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Microbiology ,Cell Line ,Viral Proteins ,Immune system ,Virology ,medicine ,Animals ,Humans ,ASK1 ,Phosphorylation ,Kinase activity ,Protein kinase A ,Immune Evasion ,Mutation ,Innate immune system ,biology ,Interleukin-8 ,Transcription Factor RelA ,Molecular biology ,Virus-Cell Interactions ,Cell biology ,Herpes simplex virus ,Insect Science ,Host-Pathogen Interactions ,biology.protein ,Mutant Proteins ,Protein Processing, Post-Translational ,Protein Binding - Abstract
Nuclear factor κB (NF-κB) plays important roles in innate immune responses by regulating the expression of a large number of target genes involved in the immune and inflammatory response, apoptosis, cell proliferation, differentiation, and survival. To survive in the host cells, viruses have evolved multiple strategies to evade and subvert the host immune response. Herpes simplex virus 1 (HSV-1) bears a large DNA genome, with the capacity to encode many different viral proteins to counteract the host immune responses. In the present study, we demonstrated that HSV-1 protein kinase US3 significantly inhibited NF-κB activation and decreased the expression of inflammatory chemokine interleukin-8 (IL-8). US3 was also shown to hyperphosphorylate p65 at serine 75 and block its nuclear translocation. Two US3 mutants, K220M and D305A, still interacted with p65; however, they could not hyperphosphorylate p65, indicating that the kinase activity of US3 was indispensable for the function. The attenuation of NF-κB activation by HSV-1 US3 protein kinase may represent a critical adaptation to enable virus persistence within the host. IMPORTANCE This study demonstrated that HSV-1 protein kinase US3 significantly inhibited NF-κB activation and decreased the expression of inflammatory chemokine interleukin-8 (IL-8). US3 hyperphosphorylated p65 at serine 75 to inhibit NF-κB activation. The kinase activity of US3 was indispensable for its hyperphosphorylation of p65 and abrogation of the nuclear translocation of p65. The present study elaborated a novel mechanism of HSV-1 US3 to evade the host innate immunity.
- Published
- 2014
- Full Text
- View/download PDF
114. PILRα and PILRβ have a siglec fold and provide the basis of binding to sialic acid
- Author
-
Guangwen Lu, Yanfang Zhang, Jinghua Yan, Jianxun Qi, George F. Gao, Zheng Fan, Chunfu Zheng, Qihui Wang, Han Wang, Yan Li, Yifang Xuan, and Qiong Lu
- Subjects
Protein Folding ,Stereochemistry ,CHO Cells ,Biology ,Crystallography, X-Ray ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cricetulus ,Protein structure ,Viral Envelope Proteins ,Animals ,Humans ,Disulfides ,Receptors, Immunologic ,Binding site ,Sialic Acid Binding Immunoglobulin-like Lectins ,Membrane Glycoproteins ,Multidisciplinary ,SIGLEC ,Biological Sciences ,N-Acetylneuraminic Acid ,Protein Structure, Tertiary ,Sialic acid ,chemistry ,Mutagenesis, Site-Directed ,Protein folding ,Protein stabilization ,N-Acetylneuraminic acid - Abstract
Paired immunoglobulin-like type 2 receptor α (PILRα) and β (PILRβ) belong to the PILR family and are related to innate immune regulation in various species. Despite their high sequence identity, PILRα and PILRβ are shown to have variant sialic acid (SA) binding avidities. To explore the molecular basis of this interaction, we solved the crystal structures of PILRα and PILRβ at resolutions of 1.6 Å and 2.2 Å, respectively. Both molecules adopt a typical siglec fold but use a hydrophobic bond to substitute the siglec-specific disulfide linkage for protein stabilization. We further used HSV-1 glycoprotein B (gB) as a representative molecule to study the PILR-SA interaction. Deploying site-directed mutagenesis, we demonstrated that three residues (Y2, R95, and W108) presented on the surface of PILRα form the SA binding site equivalent to those in siglecs but are arranged in a unique linear mode. PILRβ differs from PILRα in one of these three residues (L108), explaining its inability to engage gB. Mutation of L108 to tryptophan in PILRβ restored the gB-binding capacity. We further solved the structure of this PILRβ mutant complexed with SA, which reveals the atomic details mediating PILR/SA recognition. In comparison with the free PILR structures, amino acid Y2 oriented variantly in the complex structure, thereby disrupting the linear arrangement of PILR residues Y2, R95, and W108. In conclusion, our study provides significant implications for the PILR-SA interaction and paves the way for understanding PILR-related ligand binding.
- Published
- 2014
- Full Text
- View/download PDF
115. Herpes Simplex Virus 1 Ubiquitin-Specific Protease UL36 Inhibits Beta Interferon Production by Deubiquitinating TRAF3
- Author
-
Chunfu Zheng, Kezhen Wang, Shuai Wang, and Jie Li
- Subjects
viruses ,Immunology ,Herpesvirus 1, Human ,medicine.disease_cause ,Sendai virus ,Microbiology ,Virus ,Cell Line ,Deubiquitinating enzyme ,Viral Proteins ,Interferon ,Virology ,medicine ,Animals ,Humans ,Innate immune system ,TNF Receptor-Associated Factor 3 ,biology ,Ubiquitin ,Interferon-beta ,biology.organism_classification ,Molecular biology ,Virus-Cell Interactions ,Herpes simplex virus ,Insect Science ,Host-Pathogen Interactions ,biology.protein ,Ubiquitin-Specific Proteases ,IRF3 ,Interferon regulatory factors ,medicine.drug - Abstract
Interferon (IFN)-mediated innate immune defense is a potent antiviral mechanism. Viruses evade innate immunity and limit secretion of beta interferon (IFN-β) to replicate and survive in the host. The largest tegument protein of herpes simplex virus 1 (HSV-1), UL36, contains a novel deubiquitinase (DUB) motif embedded in its N terminus, denoted UL36 ubiquitin-specific protease (UL36USP). In the present study, we demonstrate that HSV-1 UL36USP inhibits Sendai virus (SeV)-induced interferon regulatory factor 3 (IRF3) dimerization, promoter activation, and transcription of IFN-β. The DUB activity of UL36USP is essential to block IFN-β production. UL36USP also inhibited IFN-β promoter activity induced by overexpression of the N terminus of RIG-I (RIG-IN) and MAVS, but not TBK-1, IκB kinase ε (IKKε), and IRF3/5D. UL36USP was subsequently shown to deubiquitinate TRAF3 and prevent the recruitment of the downstream adaptor TBK1. The recombinant HSV-1 lacking UL36USP DUB activity was generated. Cells infected with the mutant virus produced more IFN-β than wild-type (WT) HSV-1-infected cells. These findings demonstrate HSV-1 UL36USP removes polyubiquitin chains on TRAF3 and counteracts the IFN-β pathway.
- Published
- 2013
- Full Text
- View/download PDF
116. Herpes Simplex Virus 1 Serine/Threonine Kinase US3 Hyperphosphorylates IRF3 and Inhibits Beta Interferon Production
- Author
-
Chunfu Zheng, Kezhen Wang, Shuai Wang, and Rongtuan Lin
- Subjects
viruses ,Amino Acid Motifs ,Immunology ,Down-Regulation ,Herpesvirus 1, Human ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Microbiology ,Mice ,Viral Proteins ,Virology ,medicine ,Animals ,Humans ,Kinase activity ,Phosphorylation ,Promoter Regions, Genetic ,Serine/threonine-specific protein kinase ,Innate immune system ,Kinase ,Wild type ,virus diseases ,Herpes Simplex ,Interferon-beta ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Sendai virus ,Virus-Cell Interactions ,Mice, Inbred C57BL ,Herpes simplex virus ,Insect Science ,Female ,Interferon Regulatory Factor-3 ,IRF3 ,Protein Binding - Abstract
Viral infection initiates a series of signaling cascades that lead to the transcription of interferons (IFNs), finally inducing interferon-stimulated genes (ISGs) to eliminate viruses. Viruses have evolved a variety of strategies to modulate host IFN-mediated immune responses. Herpes simplex virus type 1 (HSV-1) US3, a Ser/Thr kinase conserved in α-herpesviruses, was previously reported to counteract the host innate immunity, however, the molecular mechanism is elusive. In this study, we report that US3 block IFN-β production by hyperphosphorylating IRF3. Ectopic expression of US3 protein significantly inhibited Sendai virus (SeV) mediated activation of the IFN-β and IFN stimulated response elements (ISRE) promoters and the transcription of IFN-β, ISG54 and ISG56. US3 was also shown to block SeV-induced dimerization and nuclear translocation of IRF3. The kinase activity was indispensable for its inhibitory function, as kinase dead (KD) US3 mutants K220M and D305A could not inhibit the IFN-β production. Furthermore, US3 interacted with and hyperphosphorylated IRF3 at Ser 175 to prevent IRF3 activation. Finally, the US3 kinase dead mutant viruses were constructed and denoted as K220M or D305A HSV-1 respectively. Cells and mice infected with both mutant viruses produced remarkable higher amount of IFN-β than wild type HSV-1. To our knowledge, it is for the first report that IRF3 is demonstrated to serve as a substrate of HSV-1 US3. These findings provide convincing evidence that US3 hyperphosphorylates IRF3, blocks the production of IFN-β, and subverts of the host innate immunity.
- Published
- 2013
- Full Text
- View/download PDF
117. Herpes Simplex Virus 1 Ubiquitin-Specific Protease UL36 Abrogates NF-κB Activation in DNA Sensing Signal Pathway
- Author
-
Ruijie Ye, Haiyan Xu, Chunfu Zheng, and Chenhe Su
- Subjects
0301 basic medicine ,Immunology ,IκB kinase ,Herpesvirus 1, Human ,Microbiology ,Deubiquitinating enzyme ,03 medical and health sciences ,chemistry.chemical_compound ,Viral Proteins ,Interferon ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Vero Cells ,Innate immune system ,biology ,NF-kappa B ,Ubiquitination ,DNA virus ,NF-κB ,Interferon-beta ,Cell biology ,Virus-Cell Interactions ,IκBα ,030104 developmental biology ,HEK293 Cells ,chemistry ,Insect Science ,Stimulator of interferon genes ,Proteolysis ,biology.protein ,medicine.drug ,Signal Transduction - Abstract
The DNA sensing pathway triggers innate immune responses against DNA virus infection, and NF-κB signaling plays a critical role in establishing innate immunity. We report here that the herpes simplex virus 1 (HSV-1) ubiquitin-specific protease (UL36USP), which is a deubiquitinase (DUB), antagonizes NF-κB activation, depending on its DUB activity. In this study, ectopically expressed UL36USP blocked promoter activation of beta interferon (IFN-β) and NF-κB induced by cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING). UL36USP restricted NF-κB activation mediated by overexpression of STING, TANK-binding kinase 1, IκB kinase α (IKKα), and IKKβ, but not p65. UL36USP was also shown to inhibit IFN-stimulatory DNA-induced IFN-β and NF-κB activation under conditions of HSV-1 infection. Furthermore, UL36USP was demonstrated to deubiquitinate IκBα and restrict its degradation and, finally, abrogate NF-κB activation. More importantly, the recombinant HSV-1 lacking UL36USP DUB activity, denoted as C40A mutant HSV-1, failed to cleave polyubiquitin chains on IκBα. For the first time, UL36USP was shown to dampen NF-κB activation in the DNA sensing signal pathway to evade host antiviral innate immunity. IMPORTANCE It has been reported that double-stranded-DNA-mediated NF-κB activation is critical for host antiviral responses. Viruses have established various strategies to evade the innate immune system. The N terminus of the HSV-1 UL36 gene-encoded protein contains the DUB domain and is conserved across all herpesviruses. This study demonstrates that UL36USP abrogates NF-κB activation by cleaving polyubiquitin chains from IκBα and therefore restricts proteasome-dependent degradation of IκBα and that DUB activity is indispensable for this process. This study expands our understanding of the mechanisms utilized by HSV-1 to evade the host antiviral innate immune defense induced by NF-κB signaling.
- Published
- 2016
118. CTCF interacts with the lytic HSV-1 genome to promote viral transcription
- Author
-
Danfeng Lu, Jumin Zhou, Chunfu Zheng, Vikrant Singh, Lihong Li, Fengchao Lang, Olga Vladimirova, Sheryl T. Smith, Xin Li, Jayamanna Wickramasinghe, Paul M. Lieberman, Benxia Hu, Nigel W. Fraser, Guijun Chen, Hongbo Han, Qihan Li, and Yu Xiao
- Subjects
0301 basic medicine ,CCCTC-Binding Factor ,Transcription, Genetic ,viruses ,RNA polymerase II ,Genome, Viral ,Herpesvirus 1, Human ,Virus Replication ,Article ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,Transcription (biology) ,Chlorocebus aethiops ,Animals ,Humans ,Author Correction ,Vero Cells ,Genetics ,Multidisciplinary ,biology ,RNA ,Chromatin Assembly and Disassembly ,Chromatin ,030104 developmental biology ,HEK293 Cells ,chemistry ,Lytic cycle ,Viral replication ,CTCF ,biology.protein ,RNA, Viral ,RNA Polymerase II ,DNA ,HeLa Cells ,Protein Binding - Abstract
CTCF is an essential chromatin regulator implicated in important nuclear processes including in nuclear organization and transcription. Herpes Simplex Virus-1 (HSV-1) is a ubiquitous human pathogen, which enters productive infection in human epithelial and many other cell types. CTCF is known to bind several sites in the HSV-1 genome during latency and reactivation, but its function has not been defined. Here, we report that CTCF interacts extensively with the HSV-1 DNA during lytic infection by ChIP-seq, and its knockdown results in the reduction of viral transcription, viral genome copy number and virus yield. CTCF knockdown led to increased H3K9me3 and H3K27me3, and a reduction of RNA pol II occupancy on viral genes. Importantly, ChIP-seq analysis revealed that there is a higher level of CTD Ser2P modified RNA Pol II near CTCF peaks relative to the Ser5P form in the viral genome. Consistent with this, CTCF knockdown reduced the Ser2P but increased Ser5P modified forms of RNA Pol II on viral genes. These results suggest that CTCF promotes HSV-1 lytic transcription by facilitating the elongation of RNA Pol II and preventing silenced chromatin on the viral genome.
- Published
- 2016
119. Herpes Simplex Virus 1 Serine Protease VP24 Blocks the DNA-Sensing Signal Pathway by Abrogating Activation of Interferon Regulatory Factor 3
- Author
-
Chenhe Su, Dandan Zhang, and Chunfu Zheng
- Subjects
0301 basic medicine ,Male ,viruses ,Immunology ,Foreskin ,Herpesvirus 1, Human ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Transfection ,Virus Replication ,Microbiology ,Cell Line ,03 medical and health sciences ,Viral Proteins ,Interferon ,Virology ,medicine ,Humans ,RNA, Small Interfering ,Promoter Regions, Genetic ,Immune Evasion ,030102 biochemistry & molecular biology ,NF-kappa B ,DNA virus ,DNA ,Interferon-beta ,Fibroblasts ,Cell biology ,Virus-Cell Interactions ,030104 developmental biology ,Herpes simplex virus ,HEK293 Cells ,Viral replication ,Insect Science ,Stimulator of interferon genes ,Gene Knockdown Techniques ,Host-Pathogen Interactions ,Interferon Regulatory Factor-3 ,Signal transduction ,IRF3 ,Interferon regulatory factors ,medicine.drug ,Signal Transduction - Abstract
The interferon (IFN)-mediated antiviral response is a central aspect of host defense; however, viruses have evolved multiple strategies to counteract IFN-mediated responses in order to successfully infect the host. Herpes simplex virus 1 (HSV-1), a typical human-restricted DNA virus, is capable of counteracting host immune responses via several distinct viral proteins, thus establishing a lifelong latent infection. In this study, we demonstrate that the VP24 protein, a serine protease of HSV-1 essential for the formation and maturation of capsids, is a novel antagonist of the beta interferon (IFN-β) pathway. Here, VP24 was shown for the first time to dampen interferon stimulatory DNA (ISD)-triggered IFN-β production and inhibit IFN-β promoter activation induced by cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) and by STING, respectively. Further study demonstrated that ectopic expression of VP24 selectively blocked IFN regulatory factor 3 (IRF3) but not NF-κB promoter activation. In addition, VP24 was demonstrated to downregulate ISD-induced phosphorylation and dimerization of IRF3 during HSV-1 infection with a VP24 stable knockdown human foreskin fibroblast cell line. The underlying molecular mechanism is that VP24 abrogates the interaction between TANK-binding kinase 1 (TBK1) and IRF3, hence impairing IRF3 activation. These results illustrate that VP24 is able to block the production of IFN-β by inhibiting IRF3 activation, which may represent a critical adaptation to enable viral effective replication within the host. IMPORTANCE This study demonstrated that HSV-1 protein VP24 could inhibit IFN-β production and promoter activation triggered by ISD, cGAS and STING and by STING, respectively. VP24 selectively blocked IRF3 promoter activation and ISD-induced phosphorylation and dimerization of IRF3 without affecting the NF-κB promoter activation during viral infection. VP24 also inhibited IRF3 activation by impeding the interaction between TBK1 and IRF3 during viral infection. This study provides new insights into the immune evasion mediated by HSV-1 and identifies VP24 as a crucial effector for HSV-1 to evade the host DNA-sensing signal pathway.
- Published
- 2016
120. The nucleolus and herpesviral usurpation
- Author
-
Shuai Wang, Liwen Ni, and Chunfu Zheng
- Subjects
Microbiology (medical) ,Cell cycle checkpoint ,Nucleolus ,Viral protein ,viruses ,cells ,genetic processes ,Ribosome biogenesis ,macromolecular substances ,Biology ,medicine.disease_cause ,environment and public health ,Microbiology ,Viral Proteins ,Stress sensing ,medicine ,Animals ,Humans ,Compartment (development) ,Herpesviridae ,Genetics ,Cell Cycle Checkpoints ,Herpesviridae Infections ,General Medicine ,Transport protein ,Cell biology ,Protein Transport ,Ribosomes ,Cell Nucleolus ,Function (biology) - Abstract
The nucleolus is a distinct subnuclear compartment known as the site for ribosome biogenesis in eukaryotes. Consequently, the nucleolus is also proposed to function in cell-cycle control, stress sensing and senescence, as well as in viral infection. An increasing number of viral proteins have been found to localize to the nucleolus. In this article, we review the current understanding of the functions of the nucleolus, the molecular mechanism of cellular and viral protein targeting to the nucleolus and the functional roles of the nucleolus during viral infection with a specific focus on the herpesvirus family.
- Published
- 2012
- Full Text
- View/download PDF
121. Herpesviral infection and Toll-like receptor 2
- Author
-
Chunfu Zheng, Meili Li, and Mingsheng Cai
- Subjects
Review ,Adaptive Immunity ,Biology ,Biochemistry ,Immune system ,Drug Discovery ,medicine ,Humans ,Herpesviridae ,Immune Evasion ,Toll-like receptor ,Innate immune system ,NF-kappa B ,Pattern recognition receptor ,Herpesviridae Infections ,Cell Biology ,Acquired immune system ,Immunity, Innate ,Toll-Like Receptor 2 ,TLR2 ,Gene Expression Regulation ,Host-Pathogen Interactions ,Interferon Regulatory Factors ,Interferon Type I ,Immunology ,Signal transduction ,Interferon type I ,Signal Transduction ,Biotechnology ,medicine.drug - Abstract
In the last decade, substantial progress has been made in understanding the molecular mechanisms involved in the initial host responses to viral infections. Herpesviral infections can provoke an inflammatory cytokine response, however, the innate pathogen-sensing mechanisms that transduce the signal for this response are poorly understood. In recent years, it has become increasingly evident that the Toll-like receptors (TLRs), which are germline-encoded pattern recognition receptors (PRRs), function as potent sensors for infection. TLRs can induce the activation of the innate immunity by recruiting specific intracellular adaptor proteins to initiate signaling pathways, which then culminating in activation of the nuclear factor kappa B (NF-κB) and interferon-regulatory factors (IRFs) that control the transcription of genes encoding type I interferon (IFN I) and other inflammatory cytokines. Furthermore, activation of innate immunity is critical for mounting adaptive immune responses. In parallel, common mechanisms used by viruses to counteract TLR-mediated responses or to actively subvert these pathways that block recognition and signaling through TLRs for their own benefit are emerging. Recent findings have demonstrated that TLR2 plays a crucial role in initiating the inflammatory process, and surprisingly that the response TLR2 triggers might be overzealous in its attempt to counter the attack by the virus. In this review, we summarize and discuss the recent advances about the specific role of TLR2 in triggering inflammatory responses in herpesvirus infection and the consequences of the alarms raised in the host that they are assigned to protect.
- Published
- 2012
- Full Text
- View/download PDF
122. The potential link between PML NBs and ICP0 in regulating lytic and latent infection of HSV-1
- Author
-
Shuai Wang, Jing Long, and Chunfu Zheng
- Subjects
Ubiquitin-Protein Ligases ,viruses ,Intranuclear Inclusion Bodies ,Human pathogen ,Herpesvirus 1, Human ,Review ,medicine.disease_cause ,Biochemistry ,Immediate-Early Proteins ,Promyelocytic leukemia protein ,Leukemia, Promyelocytic, Acute ,Drug Discovery ,medicine ,Humans ,Psychological repression ,biology ,food and beverages ,Herpes Simplex ,Cell Biology ,medicine.disease ,Virology ,Virus Latency ,Cold sore ,Leukemia ,Herpes simplex virus ,Lytic cycle ,Immunology ,biology.protein ,Stem cell ,Biotechnology - Abstract
Herpes simplex virus type 1 (HSV-1) is a common human pathogen causing cold sores and even more serious diseases. It can establish a latent stage in sensory ganglia after primary epithelial infections, and reactivate in response to stress or sunlight. Previous studies have demonstrated that viral immediate-early protein ICP0 plays a key role in regulating the balance between lytic and latent infection. Recently, It has been determined that promyelocytic leukemia (PML) nuclear bodies (NBs), small nuclear sub-structures, contribute to the repression of HSV-1 infection in the absence of functional ICP0. In this review, we discuss the fundamentals of the interaction between ICP0 and PML NBs, suggesting a potential link between PML NBs and ICP0 in regulating lytic and latent infection of HSV-1.
- Published
- 2012
- Full Text
- View/download PDF
123. Herpes Simplex Virus 1 Tegument Protein US11 Downmodulates the RLR Signaling Pathway via Direct Interaction with RIG-I and MDA-5
- Author
-
Junji Xing, Chunfu Zheng, Karen L. Mossman, Rongtuan Lin, and Shuai Wang
- Subjects
Interferon-Induced Helicase, IFIH1 ,viruses ,Immunology ,Down-Regulation ,Herpesvirus 1, Human ,medicine.disease_cause ,Microbiology ,Single-stranded binding protein ,Cell Line ,DEAD-box RNA Helicases ,Viral Proteins ,Interferon ,Virology ,medicine ,Receptors, Immunologic ,DEAD Box Protein 58 ,biology ,RIG-I ,RNA-Binding Proteins ,DNA virus ,Herpes Simplex ,Interferon-beta ,biology.organism_classification ,Molecular biology ,Sendai virus ,Protein Structure, Tertiary ,Herpes simplex virus ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,Binding domain ,medicine.drug ,Protein Binding ,Signal Transduction - Abstract
The interferon (IFN)-mediated antiviral response is a major defense of the host immune system. In order to complete their life cycle, viruses must modulate host IFN-mediated immune responses. Herpes simplex virus 1 (HSV-1) is a large DNA virus containing more than 80 genes, many of which encode proteins that are involved in virus-host interactions and show immune modulatory capabilities. In this study, we demonstrate that the US11 protein, an RNA binding tegument protein of HSV-1, is a novel antagonist of the beta IFN (IFN-β) pathway. US11 significantly inhibited Sendai virus (SeV)-induced IFN-β production, and its double-stranded RNA (dsRNA) binding domain was indispensable for this inhibition activity. Additionally, wild-type HSV-1 coinfection showed stronger inhibition than US11 mutant HSV-1 in SeV-induced IFN-β production. Coimmunoprecipitation analysis demonstrated that the US11 protein in HSV-1-infected cells interacts with endogenous RIG-I and MDA-5 through its C-terminal RNA-binding domain, which was RNA independent. Expression of US11 in both transfected and HSV-1-infected cells interferes with the interaction between MAVS and RIG-I or MDA-5. Finally, US11 dampens SeV-mediated IRF3 activation. Taken together, the combined data indicate that HSV-1 US11 binds to RIG-I and MDA-5 and inhibits their downstream signaling pathway, preventing the production of IFN-β, which may contribute to the pathogenesis of HSV-1 infection.
- Published
- 2012
- Full Text
- View/download PDF
124. Varicella-Zoster Virus Immediate-Early Protein ORF61 Abrogates the IRF3-Mediated Innate Immune Response through Degradation of Activated IRF3
- Author
-
Shuai Wang, Junji Xing, Shuping Li, Chunfu Zheng, Huifang Zhu, Karen L. Mossman, and Rongtuan Lin
- Subjects
Herpesvirus 3, Human ,Proteasome Endopeptidase Complex ,viruses ,Blotting, Western ,Immunology ,Response element ,medicine.disease_cause ,Sendai virus ,Microbiology ,Immediate early protein ,Virus ,Cell Line ,Viral Proteins ,Interferon ,Virology ,Protein Interaction Mapping ,medicine ,Humans ,Immunoprecipitation ,Immune Evasion ,Innate immune system ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Ubiquitin ,Varicella zoster virus ,virus diseases ,Interferon-beta ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Molecular biology ,Immunity, Innate ,Insect Science ,Pathogenesis and Immunity ,Interferon Regulatory Factor-3 ,IRF3 ,Protein Binding ,medicine.drug - Abstract
Varicella-zoster virus (VZV) infection of differentiated cells within the host and establishment of latency likely requires evasion of innate immunity and limits secretion of antiviral cytokines. Here we report that its immediate-early protein ORF61 antagonizes the beta interferon (IFN-β) pathway. VZV infection down-modulated the Sendai virus (SeV)-activated IFN-β pathway, including mRNA of IFN-β and its downstream interferon-stimulated genes (ISGs), ISG54 and ISG56. Through a primary screening of VZV genes, we found that ORF61 inhibited SeV-mediated activation of IFN-β and ISRE (IFN-stimulated response element) promoter activities but only slightly affected NF-κB promoter activity, implying that the IFN-β pathway may be blocked in the IRF3 branch. An indirect immunofluorescence assay demonstrated that ectopic expression of ORF61 abrogated the detection of IRF3 in SeV-infected cells; however, it did not affect endogenous dormant IRF3 in noninfected cells. Additionally, ORF61 was shown to be partially colocalized with activated IRF3 in the nucleus upon treatment with MG132, an inhibitor of proteasomes, and the direct interaction between ORF61 and activated IRF3 was confirmed by a coimmunoprecipitation assay. Furthermore, Western blot analysis demonstrated that activated IRF3 was ubiquitinated in the presence of ORF61, suggesting that ORF61 degraded phosphorylated IRF3 via a ubiquitin-proteasome pathway. Semiquantitative reverse transcription-PCR (RT-PCR) analysis demonstrated that the level of ISG54 and ISG56 mRNAs was also downregulated by ORF61. Taken together, our results convincingly demonstrate that ORF61 down-modulates the IRF3-mediated IFN-β pathway by degradation of activated IRF3 via direct interaction, which may contribute to the pathogenesis of VZV infection.
- Published
- 2011
- Full Text
- View/download PDF
125. A novel virus-encoded nucleocytoplasmic shuttling protein: The UL3 protein of herpes simplex virus type 1
- Author
-
Fusen Lin, Shuai Wang, Chunfu Zheng, and Junji Xing
- Subjects
Cytoplasm ,Recombinant Fusion Proteins ,viruses ,Blotting, Western ,Molecular Sequence Data ,Nuclear Localization Signals ,Active Transport, Cell Nucleus ,Receptors, Cytoplasmic and Nuclear ,Herpesvirus 1, Human ,Importin ,Karyopherins ,Biology ,Mice ,Viral Proteins ,Virology ,Chlorocebus aethiops ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Nuclear protein ,Nuclear export signal ,Sequence Deletion ,Cell Nucleus ,Nuclear Export Signals ,Subcellular localization ,Molecular biology ,Cell biology ,Microscopy, Fluorescence ,COS Cells ,Chromosomal region ,Ran ,Fatty Acids, Unsaturated ,NIH 3T3 Cells ,Nuclear localization sequence ,Plasmids - Abstract
Herpes simplex virus type 1 (HSV-1) UL3 protein is a nuclear protein. In this study, the molecular mechanism of the subcellular localization of UL3 was characterized by fluorescence microscopy in living cells. A nuclear localization signal (NLS) and a nuclear export signal (NES) were also identified. UL3 was demonstrated to target to the cytoplasm through the NES via chromosomal region maintenance 1 (CRM-1) dependent pathway, and to the nucleus through RanGTP-dependent mechanism. Heterokaryon assays confirmed that UL3 was capable of shuttling between the nucleus and the cytoplasm. These results demonstrate that the UL3 protein is a novel HSV-1 encoded nucleocytoplasmic shuttling protein.
- Published
- 2011
- Full Text
- View/download PDF
126. Identification of Nuclear and Nucleolar Localization Signals of Pseudorabies Virus (PRV) Early Protein UL54 Reveals that Its Nuclear Targeting Is Required for Efficient Production of PRV
- Author
-
Chunfu Zheng, Mingsheng Cai, Shuai Wang, and Meili Li
- Subjects
viruses ,Immunology ,Protein Sorting Signals ,Biology ,Virus Replication ,medicine.disease_cause ,Recombinant virus ,Microbiology ,Virus ,Cell Line ,Viral Proteins ,Virology ,Chlorocebus aethiops ,Gene expression ,medicine ,Animals ,Humans ,NLS ,Sequence Deletion ,Mutation ,Herpesvirus 1, Suid ,Recombinant Proteins ,Genome Replication and Regulation of Viral Gene Expression ,Protein Transport ,Herpes simplex virus ,Viral replication ,Insect Science ,Mutant Proteins ,Cell Nucleolus ,Nuclear localization sequence - Abstract
The pseudorabies virus (PRV) early protein UL54 is a homologue of herpes simplex virus 1 (HSV-1) immediate-early protein ICP27, which is a multifunctional protein that is essential for HSV-1 infection. In this study, the subcellular localization and nuclear import signals of PRV UL54 were characterized. UL54 was shown to predominantly localize to the nucleolus in transfected cells. By constructing a series of mutants, a functional nuclear localization signal (NLS) and a genuine nucleolar localization signal (NoLS) of UL54 were for the first time identified and mapped to amino acids 61 RQRRR 65 and 45 RRRRGGRGGRAAR 57 , respectively. Additionally, three recombinant viruses with mutations of the NLS and/or the NoLS in UL54 were constructed based on PRV bacterial artificial chromosome (BAC) pBecker2 to test the effect of UL54 nuclear targeting on viral replication. In comparison with the wild-type virus, a recombinant virus harboring an NLS or NoLS mutation of UL54 reduced viral production to different extents. However, mutations of both the NLS and NoLS targeted UL54 to the cytoplasm in recombinant virus-infected cells and significantly impaired viral replication, comparable to the UL54-null virus. In addition, a virus lacking the NLS or the NoLS displayed modest defects in viral gene expression and DNA synthesis. However, deletion of both the NLS and the NoLS resulted in severe defects in viral gene expression and DNA synthesis, as well as production of infectious progeny. Thus, we have identified a classical NLS and a genuine NoLS in UL54 and demonstrate that the nuclear targeting of UL54 is required for efficient production of PRV. Pseudorabies virus (PRV), a typical alphaherpesvirus, is an economically important pathogen of swine. The neurotropic nature of PRV makes it a useful tracer of neuronal connections, and it is also a useful model for the study of herpesvirus pathogenesis (44). During the lytic cycle of PRV infection, the viral proteins are expressed in a cascade of three temporally distinct and functionally interdependent phases termed immediate-early (IE), early (E), and late (L) phase. It has been reported that UL54 is expressed with E kinetics (24), whereas its homologue from herpes simplex virus 1 (HSV-1), ICP27, belongs to the IE kinetic class. Furthermore, the amino acid sequence similarity between ICP27 and UL54 is only 41% (2). ICP27 is an essential protein and a multifunctional regulator of gene expression of HSV-1, given its different roles during infection (27, 37, 45). In contrast to ICP27-null HSV-1, UL54null PRV is viable; however, it exhibits aberrant expression of several E and L gene products and is highly attenuated in a mouse model of PRV infection (47). Like its HSV-1 counterpart, UL54 contains a putative nuclear localization signal (NLS) and an RGG box RNA binding motif and shows RNA binding activity (25). All of these properties of UL54 make it of particular interest. Although ICP27 (27, 37, 45) and other
- Published
- 2011
- Full Text
- View/download PDF
127. Molecular determinants responsible for the subcellular localization of HSV-1 UL4 protein
- Author
-
Chunfu Zheng, Wei Wei Pan, Junji Xing, and Jing Long
- Subjects
Yellow fluorescent protein ,Cytoplasm ,Recombinant Fusion Proteins ,Immunology ,Active Transport, Cell Nucleus ,Herpesvirus 1, Human ,Protein Sorting Signals ,Virus Replication ,Article ,Viral Proteins ,Bacterial Proteins ,Genes, Reporter ,Virology ,Fluorescence microscope ,Animals ,Nuclear export signal ,Sequence Deletion ,Cell Nucleus ,chemistry.chemical_classification ,Staining and Labeling ,biology ,Subcellular localization ,Amino acid ,Luminescent Proteins ,Microscopy, Fluorescence ,Biochemistry ,chemistry ,Chromosomal region ,Ran ,biology.protein ,Molecular Medicine - Abstract
The function of the herpes simplex virus type 1 (HSV-1) UL4 protein is still elusive. Our objective is to investigate the subcellular transport mechanism of the UL4 protein. In this study, fluorescence microscopy was employed to investigate the subcellular localization of UL4 and characterize the transport mechanism in living cells. By constructing a series of deletion mutants fused with enhanced yellow fluorescent protein (EYFP), the nuclear export signals (NES) of UL4 were for the first time mapped to amino acid residues 178 to 186. In addition, the N-terminal 19 amino acids are identified to be required for the granule-like cytoplasmic pattern of UL4. Furthermore, the UL4 protein was demonstrated to be exported to the cytoplasm through the NES in a chromosomal region maintenance 1 (CRM1)-dependent manner involving RanGTP hydrolysis.
- Published
- 2011
- Full Text
- View/download PDF
128. Characterization of molecular determinants for nucleocytoplasmic shuttling of PRV UL54
- Author
-
Shuai Wang, Meili Li, Chunfu Zheng, Hong Guo, and Mingsheng Cai
- Subjects
Cytoplasm ,viruses ,Importin ,Biology ,TAP/NXF1 ,NXF1 ,Viral Proteins ,Virology ,Chlorocebus aethiops ,Protein Interaction Mapping ,Animals ,Nuclear protein ,Nuclear export signal ,Cell Nucleus ,Herpesvirus 1, Suid ,Molecular biology ,Transport protein ,Protein Transport ,PRV UL54 ,Ran-GTP ,COS Cells ,Ran ,Nucleocytoplasmic shuttling ,Nuclear transport - Abstract
The pseudorabies virus (PRV) early protein UL54 is a homologue of the herpes simplex virus 1 (HSV-1) immediate-early protein ICP27, which is a multifunctional protein and essential for HSV-1 infection. To determine if UL54 might shuttle between the nucleus and cytoplasm, as has been shown for its homologues in human herpesviruses, the molecular determinants for its nucleocytoplasmic shuttling were investigated. Heterokaryon assays demonstrated that UL54 was a nucleocytoplasmic shuttling protein and this property could not be blocked by leptomycin B, an inhibitor of chromosome region maintenance 1 (CRM1). However, TAP/NXF1 promoted the nuclear export of UL54 and interacted with UL54, suggesting that UL54 shuttles between the nucleus and the cytoplasm via a TAP/NXF1, but not CRM1, dependent nuclear export pathway. Furthermore, UL54 was demonstrated to target to the nucleus through a classic Ran-, importin β1- and α5-dependent nuclear import mechanism.
- Published
- 2011
- Full Text
- View/download PDF
129. Screening and identification of host factors interacting with UL14 of herpes simplex virus 1
- Author
-
Shuai Wang, Meili Li, Junji Xing, Chunfu Zheng, and Fuqing Wu
- Subjects
Microbiology (medical) ,Arrestins ,Ubiquitin-Protein Ligases ,Two-hybrid screening ,Immunology ,RNA polymerase II ,Herpesvirus 1, Human ,Virus Replication ,medicine.disease_cause ,Viral Proteins ,Transcription (biology) ,Two-Hybrid System Techniques ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Immunoprecipitation ,Immunology and Allergy ,Fluorescent Antibody Technique, Indirect ,Vero Cells ,Transcription factor ,beta-Arrestins ,Adaptor Proteins, Signal Transducing ,Mediator Complex ,biology ,Beta-Arrestins ,Cereblon ,General Medicine ,beta-Arrestin 2 ,Molecular biology ,Core Binding Factor Alpha 3 Subunit ,HEK293 Cells ,Herpes simplex virus ,Viral replication ,COS Cells ,Host-Pathogen Interactions ,biology.protein ,Peptide Hydrolases ,Plasmids ,Transcription Factors - Abstract
The UL14 protein of herpes simplex virus type 1 (HSV-1) is highly conserved in herpesvirus family. However, its exact function during the HSV-1 replication cycle is little known. In the present study, a high throughput yeast two-hybrid system was employed to screen the cellular factors interacting with UL14, and five target candidates were yielded: (1) TSC22 domain family protein 3 (TSC22D3); (2) Mediator of RNA polymerase II transcription subunit 8 isoform 1(MED8); (3) Runt-related transcription factor 3 (RUNX3); (4) Arrestin beta-2 (ARRB2); (5) Cereblon (CRBN). Indirect immunofluorescent assay showed that both TSC22D3 and MED8 co-localized with UL14. Co-immunoprecipitation assay demonstrated that UL14 could be immunoprecipitated by TSC22D3, suggesting that UL14 interacted with TSC22D3 under physiological condition. In summary, this study opened up new avenues toward delineating the function and physiological significance of UL14 during the HSV-1 replication cycle.
- Published
- 2011
- Full Text
- View/download PDF
130. Host cell targets of tegument protein VP22 of herpes simplex virus 1
- Author
-
Lin Wang, Chunfu Zheng, Meili Li, and Xiaoming Ren
- Subjects
Yellow fluorescent protein ,Gene Expression ,Herpesvirus 1, Human ,Plasma protein binding ,medicine.disease_cause ,Virus ,Two-Hybrid System Techniques ,Virology ,medicine ,Humans ,Fluorescent Dyes ,Viral Structural Proteins ,biology ,cDNA library ,General Medicine ,Viral tegument ,Subcellular localization ,Molecular biology ,High-Throughput Screening Assays ,Herpes simplex virus ,Host-Pathogen Interactions ,Ring finger protein 10 ,biology.protein ,Carrier Proteins ,Protein Binding - Abstract
In the present study, seven novel host cell factors associated with VP22 were identified from the human leukocyte cDNA library using a yeast two-hybrid high-throughput screening system. To confirm some of the interactions, VP22 and its homologues and two candidate targets were tagged with enhanced cyan or yellow fluorescent protein. While RING finger protein 10 (RNF10), WD repeat-containing protein 42A (WDR42A) or VP22 alone showed distinct subcellular localization patterns, RNF10 and WDR42A were relocated when co-expressed with VP22 or its homologues. Thus, these potential host cell factors of VP22 might expand the list of the host targets of VP22.
- Published
- 2011
- Full Text
- View/download PDF
131. Granulysin Production and Anticryptococcal Activity Is Dependent upon a Far Upstream Enhancer That Binds STAT5 in Human Peripheral Blood CD4+ T Cells
- Author
-
Chunfu Zheng, Christopher H. Mody, Shuai Wang, Alan M. Krensky, Fuqing Wu, and Junji Xing
- Subjects
Antigens, Differentiation, T-Lymphocyte ,CD4-Positive T-Lymphocytes ,Interleukin 2 ,Chromatin Immunoprecipitation ,Blotting, Western ,Immunology ,Gene Expression ,Electrophoretic Mobility Shift Assay ,Biology ,Lymphocyte Activation ,Transfection ,Article ,STAT5 Transcription Factor ,medicine ,Humans ,Immunology and Allergy ,Binding site ,Granulysin ,Promoter Regions, Genetic ,Enhancer ,Reverse Transcriptase Polymerase Chain Reaction ,food and beverages ,Molecular biology ,Upstream Enhancer ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Granulysin production ,Cryptococcus neoformans ,Chromatin immunoprecipitation ,Signal Transduction ,medicine.drug - Abstract
Previous studies have demonstrated that STAT5 is critical for expression of granulysin and antimicrobial activity. Because the signaling pathway and the resultant microbicidal activity are defective in HIV-infected patients, the mechanism by which STAT5 leads to granulysin expression is of great interest. In the current study, IL-2–stimulated CRL-2105 CD4+ T cells expressed granulysin and killed Cryptococcus neoformans similar to primary CD4+ T cells. The enhancer activity of the upstream element of the granulysin promoter was analyzed in primary CD4+ T cells and CRL-2105 T cells with a luciferase reporter assay, and a STAT5 binding site, 18,302 to 18,177 bp upstream of the transcription start site, was identified as an enhancer. Additionally, the enhancer functioned in the context of heterologous SV40 promoter irrespective of its transcriptional orientation. Chromatin immunoprecipitation and EMSAs demonstrated that the enhancer element bound STAT5 both in vivo and in vitro, and mutation of the STAT5 binding site abrogated its enhancer activity. Furthermore, overexpression of a dominant negative STAT5a abolished the enhancer activity of the STAT5 binding site and abrogated the anticryptococcal activity of IL-2–stimulated primary CD4+ T cells. Taken together, these data provide details about the complex regulation leading to granulysin expression and anticryptococcal activity in primary CD4+ T cells.
- Published
- 2010
- Full Text
- View/download PDF
132. Molecular anatomy of subcellular localization of HSV-1 tegument protein US11 in living cells
- Author
-
Junji Xing, Weiwei Pan, Chunfu Zheng, and Fuqing Wu
- Subjects
Yellow fluorescent protein ,Cancer Research ,Nucleolus ,Recombinant Fusion Proteins ,viruses ,Molecular Sequence Data ,Herpesvirus 1, Human ,Importin ,Protein Sorting Signals ,environment and public health ,Article ,Viral Proteins ,Bacterial Proteins ,Virology ,Chlorocebus aethiops ,Animals ,Amino Acid Sequence ,Herpes simplex virus 1 US11 protein ,Nuclear export signal ,Vero Cells ,Sequence Deletion ,integumentary system ,biology ,RNA-Binding Proteins ,Sequence Analysis, DNA ,Subcellular localization ,Nucleolar localization signal ,Artificial Gene Fusion ,Luminescent Proteins ,Protein Transport ,Infectious Diseases ,Microscopy, Fluorescence ,Biochemistry ,Ran-GTP ,Ran ,Chromosomal region ,biology.protein ,Nuclear transport ,Sequence Alignment - Abstract
The herpes simplex virus type I (HSV-1) US11 protein is an RNA-binding multifunctional regulator that specifically and stably associates with nucleoli. Although the C-terminal part of US11 was responsible for its nucleolar localization, the precise nucleolar localization signal (NoLS) and nuclear export signal (NES) of US11 and its nuclear import and export mechanisms are still elusive. In this study, fluorescence microscopy was employed to investigate the subcellular localization of US11 and characterize its transport mechanism in living cells. By constructing a series of deletion mutants fused with enhanced yellow fluorescent protein (EYFP), three novel NoLSs of US11 were for the first time mapped to amino acids 84–125, 126–152, and 89–146, respectively. Additionally, the NES was identified to locate between amino acids 89 and 119. Furthermore, the US11 protein was demonstrated to target to the cytoplasm through the NES by chromosomal region maintenance 1 (CRM1)-independent pathway, and to the nucleolus through Ran and importin β-dependent mechanism that does not require importin α5.
- Published
- 2010
- Full Text
- View/download PDF
133. Real-time imaging of trapping and urease-dependent transmigration of Cryptococcus neoformans in mouse brain
- Author
-
Paul Kubes, Shu Shun Li, Kwang Sik Kim, Hong Zhou, Christopher H. Mody, Chunfu Zheng, Gareth Jones, and Meiqing Shi
- Subjects
Virulence ,Models, Biological ,Exocytosis ,Microbiology ,Mice ,Phagocytosis ,Parenchyma ,medicine ,Animals ,Humans ,Pathogen ,Cryptococcus neoformans ,Microscopy ,biology ,Macrophages ,Brain ,Meningoencephalitis ,Cryptococcosis ,General Medicine ,medicine.disease ,biology.organism_classification ,Capillaries ,Cerebrovascular Circulation ,Encephalitis ,Intravital microscopy ,Research Article - Abstract
Infectious meningitis and encephalitis is caused by invasion of circulating pathogens into the brain. It is unknown how the circulating pathogens dynamically interact with brain endothelium under shear stress, leading to invasion into the brain. Here, using intravital microscopy, we have shown that Cryptococcus neoformans, a yeast pathogen that causes meningoencephalitis, stops suddenly in mouse brain capillaries of a similar or smaller diameter than the organism, in the same manner and with the same kinetics as polystyrene microspheres, without rolling and tethering to the endothelial surface. Trapping of the yeast pathogen in the mouse brain was not affected by viability or known virulence factors. After stopping in the brain, C. neoformans was seen to cross the capillary wall in real time. In contrast to trapping, viability, but not replication, was essential for the organism to cross the brain microvasculature. Using a knockout strain of C. neoformans, we demonstrated that transmigration into the mouse brain is urease dependent. To determine whether this could be amenable to therapy, we used the urease inhibitor flurofamide. Flurofamide ameliorated infection of the mouse brain by reducing transmigration into the brain. Together, these results suggest that C. neoformans is mechanically trapped in the brain capillary, which may not be amenable to pharmacotherapy, but actively transmigrates to the brain parenchyma with contributions from urease, suggesting that a therapeutic strategy aimed at inhibiting this enzyme could help prevent meningitis and encephalitis caused by C. neoformans infection.
- Published
- 2010
- Full Text
- View/download PDF
134. A multiple functional protein: the herpes simplex virus type 1 tegument protein VP22
- Author
-
Mei-li Li, Chunfu Zheng, Hong Guo, and Qiong Ding
- Subjects
chemistry.chemical_classification ,biology ,viruses ,Chromatin binding ,Intercellular transport ,Immunology ,Viral tegument ,medicine.disease_cause ,Molecular biology ,Single-stranded binding protein ,Herpes simplex virus ,chemistry ,Microtubule ,Virology ,medicine ,biology.protein ,Protein biosynthesis ,Molecular Medicine ,Glycoprotein - Abstract
The herpes simplex virus type 1 (HSV-1) VP22, is one of the most abundant HSV-1 tegument proteins with an average stoichiometry of 2 400 copies per virion and conserved among alphaherpesvirinae. Many functions are attributed to VP22, including nuclear localization, chromatin binding, microtubule binding, induction of microtubule reorganization, intercellular transport, interaction with cellular proteins, such as template activating factor I (TAF-I) and nonmuscle myosin II A (NMIIA), and viral proteins including tegument protein VP16, pUS9 and pUL46, glycoprotein E (gE) and gD. Recently, many novel functions performed by the HSV-1 VP22 protein have been shown, including promotion of protein synthesis at late times in infection, accumulation of a subset of viral mRNAs at early times in infection and possible transcriptional regulation function.
- Published
- 2009
- Full Text
- View/download PDF
135. The herpes simplex virus type 1 multiple function protein ICP27
- Author
-
Gui-qing Peng, Qiong Ding, Chunfu Zheng, Lei Zhao, and Wen-bo Zhu
- Subjects
Regulation of gene expression ,viruses ,Immunology ,Host gene ,Biology ,medicine.disease_cause ,Virology ,Herpes simplex virus ,Multiple function ,RNA splicing ,Leptomycin B ,medicine ,Molecular Medicine ,Nuclear export signal ,Precursor mRNA - Abstract
The herpes simplex virus type 1 (HSV-1) infected-cell protein 27 (ICP27) is an essential, highly conserved protein involved in various steps of HSV-1 gene regulation as well as in the shut-off of host gene expression during infection. It functions primarily at the post-transcriptional level in inhibiting precursor mRNA splicing and in promoting nuclear export of viral transcripts. Recently, many novel functions performed by the HSV-1 ICP27 protein were shown, including leptomycin B resistance, inhibition of the typeⅠinterferon signaling, regulation of the viral mRNA translation and determining the composition of HSV-1 virions.
- Published
- 2008
- Full Text
- View/download PDF
136. Late Expression of Granulysin by Microbicidal CD4+ T Cells Requires PI3K- and STAT5-Dependent Expression of IL-2Rβ That Is Defective in HIV-Infected Patients
- Author
-
Chunfu Zheng, M. John Gill, Meiqing Shi, Shaunna M. Huston, Christopher H. Mody, Alan M. Krensky, Gareth Jones, Byron M. Berenger, Kaleb J. Marr, Paul Kubes, and Jeremy C. D. Wiseman
- Subjects
Antigens, Differentiation, T-Lymphocyte ,CD4-Positive T-Lymphocytes ,Interleukin 2 ,Small interfering RNA ,T cell ,Immunology ,HIV Infections ,Biology ,Article ,Phosphatidylinositol 3-Kinases ,STAT5 Transcription Factor ,medicine ,Class Ib Phosphatidylinositol 3-Kinase ,Humans ,Immunology and Allergy ,Enzyme Inhibitors ,Granulysin ,STAT5 ,Phosphoinositide-3 Kinase Inhibitors ,Cell biology ,Interleukin-2 Receptor beta Subunit ,Isoenzymes ,Cytolysis ,medicine.anatomical_structure ,Granulysin production ,Cryptococcus neoformans ,biology.protein ,Interleukin-2 ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
Granulysin is a cytolytic effector molecule used by lymphocytes to kill tumor and microbial cells. Regulation of granulysin production is complex. A significant delay (5 days) following stimulation of CD4+ T cells with IL-2 occurs before granulysin is produced. Unfortunately, the mechanisms responsible for this delay are unknown. We have recently demonstrated that granulysin-mediated killing of Cryptococcus neoformans by CD4+ T cells is defective during HIV infection. This is because CD4+ T cells from HIV-infected patients fail to produce granulysin in response to IL-2 activation. The present studies examined the mechanism of delayed production of granulysin and the mechanism of the defect in HIV patients. We demonstrate that IL-2 initially requires both STAT5 and PI3K activation to increase expression of IL-2Rβ, produce granulysin, and kill C. neoformans. The increased expression of IL-2Rβ precedes granulysin, and preventing the increased expression of IL-2Rβ using small interfering RNA knockdown abrogates granulysin expression. Moreover, following the increased expression of IL-2Rβ, blocking subsequent signaling by IL-2 using IL-2Rβ-specific blocking Abs abrogates expression of granulysin. Finally, CD4+ T cells from HIV-infected patients, who are defective in both STAT5 and PI3K signaling, fail to express IL-2Rβ and fail to produce granulysin. These results suggest that IL-2 signals via PI3K and STAT5 to increase expression of IL-2Rβ, which in turn is required for production of granulysin. These results provide a mechanism to explain the “late” production of granulysin during normal T cell responses, as well as for defective granulysin production by CD4+ T cells in HIV-infected patients.
- Published
- 2008
- Full Text
- View/download PDF
137. Herpes simplex virus 1 UL41 protein abrogates the antiviral activity of hZAP by degrading its mRNA
- Author
-
Jie Zhang, Chunfu Zheng, and Chenhe Su
- Subjects
RNA Stability ,Subfamily ,viruses ,RNA-binding protein ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Cell Line ,Viral Proteins ,Virology ,medicine ,Animals ,Humans ,Messenger RNA ,UL41 ,Immune evasion ,Research ,RNA-Binding Proteins ,hZAP ,HSV-1 ,Infectious Diseases ,Herpes simplex virus ,Cell culture ,Host-Pathogen Interactions ,Molecular mechanism ,Ectopic expression - Abstract
Background The zinc finger antiviral protein (ZAP) is a host restriction factor that inhibits the replication of various viruses by degradation of certain viral mRNA. However, previous study demonstrated that ectopic expression of rat ZAP did not suppress the replication of herpes simplex virus type 1 (HSV-1), an archetypal member of the alphaherpesvirus subfamily, and the molecular mechanism underneath is still illusive. Results Human ZAP (hZAP) does not suppress the replication of herpes simplex virus 1, and HSV-1 UL41 protein was identified as an antagonist of hZAP by degrading its mRNA. Infection of wild-type (WT), but not UL41-null mutant (R2621) virus, diminished the accumulation of hZAP to abrogate its antiviral activity. Moreover, ectopic expression of hZAP inhibited the replication of R2621 but not WT HSV-1. Conclusion HSV-1 UL41 was shown for the first time to evade the antiviral function of hZAP via its RNase activity.
- Published
- 2015
- Full Text
- View/download PDF
138. Zinc finger antiviral protein inhibits coxsackievirus B3 virus replication and protects against viral myocarditis
- Author
-
Lin Wei, Chunfu Zheng, Jie Yang, Chenyu Lu, Fei Xiong, Min Li, Kepeng Yan, and Wei Xu
- Subjects
Male ,Viral Myocarditis ,Myocarditis ,viruses ,Coxsackievirus Infections ,chemical and pharmacologic phenomena ,Filoviridae ,medicine.disease_cause ,Virus Replication ,Virology ,medicine ,Animals ,Humans ,Pharmacology ,Mice, Inbred BALB C ,biology ,Gene Expression Profiling ,Myocardium ,virus diseases ,RNA ,RNA-Binding Proteins ,hemic and immune systems ,RNA virus ,biology.organism_classification ,medicine.disease ,Immunity, Innate ,Enterovirus B, Human ,Disease Models, Animal ,Viral replication ,Togaviridae ,Enterovirus ,Cytokines ,RNA, Viral ,HeLa Cells ,Protein Binding - Abstract
The host Zinc finger antiviral protein (ZAP) has been reported exhibiting antiviral activity against positive-stranded RNA viruses (Togaviridae), negative-stranded RNA viruses (Filoviridae) and retroviruses (Retroviridae). However, whether ZAP restricts the infection of enterovirus and the development of enterovirus mediated disease remains unknown. Here, we reported the antiviral properties of ZAP against coxsackievirus B3 (CVB3), a single-stranded RNA virus of the Enterovirus genus within the Picornaviridae as a major causative agent of viral myocarditis (VMC). We found that the expression of ZAP was significantly induced after CVB3 infection in heart tissues of VMC mice. ZAP potently inhibited CVB3 replication in cells after infection, while overexpression of ZAP in mice significantly increased the resistance to CVB3 replication and viral myocarditis by significantly reducing cardiac inflammatory cytokine production. The ZAP-responsive elements (ZREs) were mapped to the 3'UTR and 5'UTR of viral RNA. Taken together, ZAP confers resistance to CVB3 infection via directly targeting viral RNA and protects mice from acute myocarditis by suppressing viral replication and cardiac inflammatory cytokine production. Our finding further expands ZAP's range of viral targets, and suggests ZAP as a potential therapeutic target for viral myocarditis caused by CVB3.
- Published
- 2015
139. Intercellular trafficking of the major tegument protein VP22 of bovine herpesvirus-1 and its application to improve a DNA vaccine
- Author
-
Chunfu Zheng, Lorne A. Babiuk, D. Y. Huang, S. van Drunen Littel-van den Hurk, and Robert Brownlie
- Subjects
Recombinant Fusion Proteins ,T-Lymphocytes ,Green Fluorescent Proteins ,Antibodies, Viral ,Virus ,Interferon-gamma ,Mice ,Viral Proteins ,Immune system ,Plasmid ,Genes, Reporter ,Virology ,Alphaherpesvirinae ,Vaccines, DNA ,Animals ,Herpesvirus 1, Bovine ,Viral Structural Proteins ,chemistry.chemical_classification ,biology ,Viral Vaccines ,General Medicine ,Viral tegument ,Transfection ,biology.organism_classification ,Molecular biology ,Bovine herpesvirus 1 ,Amino acid ,chemistry ,Immunoglobulin G ,Interleukin-4 ,Plasmids - Abstract
Intercellular spread of bovine herpesvirus-1 (BHV-1) VP22 was demonstrated in living COS-7 cells transfected with a plasmid expressing VP22-YFP (yellow fluorescence protein) and CFP (cyan fluorescence protein) bicistronically. The intercellular trafficking property of VP22 was localized to the C-terminal portion of the molecule (amino acids 121-258; VP22-C). Plasmids encoding a truncated form of BHV-1 glycoprotein D (tgD) fused to VP22, VP22-C, or the N-terminal portion of VP22 (amino acids 1-120; VP22-N) were constructed. Mice immunized with plasmid encoding tgD-VP22 or tgD-VP22-C developed stronger immune responses when compared to animals immunized with plasmid encoding tgD or tgD fused to tgD-VP22-N.
- Published
- 2005
- Full Text
- View/download PDF
140. Characterization of the Nuclear Localization and Nuclear Export Signals of Bovine Herpesvirus 1 VP22
- Author
-
Chunfu Zheng, Lorne A. Babiuk, Sylvia van Drunen Littel-van den Hurk, and Robert Brownlie
- Subjects
Recombinant Fusion Proteins ,viruses ,Amino Acid Motifs ,Nuclear Localization Signals ,Immunology ,Active Transport, Cell Nucleus ,Protein Sorting Signals ,Biology ,Microbiology ,Cell Line ,chemistry.chemical_compound ,Bacterial Proteins ,Virology ,Chlorocebus aethiops ,Animals ,NLS ,Amino Acid Sequence ,Nuclear export signal ,Peptide sequence ,Herpesvirus 1, Bovine ,Viral Structural Proteins ,chemistry.chemical_classification ,Base Sequence ,Leptomycin ,Subcellular localization ,Molecular biology ,Genome Replication and Regulation of Viral Gene Expression ,Amino acid ,Luminescent Proteins ,chemistry ,Cytoplasm ,Insect Science ,COS Cells ,DNA, Viral ,Cattle ,Nuclear localization sequence ,Subcellular Fractions - Abstract
The bovine herpesvirus 1 (BHV-1) tegument protein VP22 is predominantly localized in the nucleus after viral infection. To analyze subcellular localization in the absence of other viral proteins, a plasmid expressing BHV-1 VP22 fused to enhanced yellow fluorescent protein (EYFP) was constructed. The transient expression of VP22 fused to EYFP in COS-7 cells confirmed the predominant nuclear localization of VP22. Analysis of the amino acid sequence of VP22 revealed that it does not have a classical nuclear localization signal (NLS). However, by constructing a series of deletion derivatives, we mapped the nuclear targeting domain of BHV-1 VP22 to amino acids (aa) 121 to 139. Furthermore, a 4-aa motif, 130 PRPR 133 , was able to direct EYFP and an EYFP dimer (dEYFP) or trimer (tEYFP) predominantly into the nucleus, whereas a deletion or mutation of this arginine-rich motif abrogated the nuclear localization property of VP22. Thus, 130 PRPR 133 is a functional nonclassical NLS. Since we observed that the C-terminal 68 aa of VP22 mediated the cytoplasmic localization of EYFP, an analysis was performed on these C-terminal amino acid sequences, and a leucine-rich motif, 204 LDRMLKSAAIRIL 216 , was detected. Replacement of the leucines in this putative nuclear export signal (NES) with neutral amino acids resulted in an exclusive nuclear localization of VP22. Furthermore, this motif was able to localize EYFP and dEYFP in the cytoplasm, and the nuclear export function of this NES could be blocked by leptomycin B. This demonstrates that this leucine-rich motif is a functional NES. These data represent the first identification of a functional NLS and NES in a herpesvirus VP22 homologue.
- Published
- 2005
- Full Text
- View/download PDF
141. Bovine Herpesvirus 1 VP22 Enhances the Efficacy of a DNA Vaccine in Cattle
- Author
-
Sylvia van Drunen Littel-van den Hurk, Chunfu Zheng, and Lorne A. Babiuk
- Subjects
Recombinant Fusion Proteins ,Immunology ,Cattle Diseases ,Context (language use) ,Herpesvirus Vaccines ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Herpesviridae ,Virus ,Viral Proteins ,Plasmid ,Immune system ,Adjuvants, Immunologic ,Immunity ,Virology ,Alphaherpesvirinae ,Vaccines and Antiviral Agents ,Vaccines, DNA ,medicine ,Animals ,Herpesvirus 1, Bovine ,Viral Structural Proteins ,biology ,Herpesviridae Infections ,biology.organism_classification ,Bovine herpesvirus 1 ,Insect Science ,COS Cells ,Cattle ,Immunization - Abstract
For this study, the intercellular trafficking ability of bovine herpesvirus 1 (BHV-1) VP22 was applied to improve the efficacy of a DNA vaccine in calves. A plasmid encoding a truncated version of glycoprotein D (tgD) fused to VP22 was constructed. The plasmid encoding tgD-VP22 elicited significantly enhanced and more balanced immune responses than those induced by a plasmid encoding tgD. Furthermore, protection against a BHV-1 challenge was obtained in calves immunized with the plasmid encoding tgD-VP22, as shown by significant reductions in viral excretion. However, less significant protection was observed for animals vaccinated with the tgD-expressing plasmid, correlating with the lower level of immunity observed prechallenge. This is the first report of the use of VP22 as a transport molecule in the context of a DNA vaccine for a large animal species.
- Published
- 2005
- Full Text
- View/download PDF
142. Molecular Cloning and Sequencing of the Merozoite Surface Antigen 2 Gene from Plasmodium falciparum Strain FCC-l/HN and Expression of the Gene in Mycobacteria1
- Author
-
Peimei Xie, Chunfu Zheng, and Yatang Chen
- Subjects
Genetic Vectors ,Molecular Sequence Data ,Plasmodium falciparum ,Protozoan Proteins ,Antigens, Protozoan ,Biology ,Molecular cloning ,Microbiology ,Mycobacterium tuberculosis ,Plasmid ,Malaria Vaccines ,Animals ,Humans ,HSP70 Heat-Shock Proteins ,Amino Acid Sequence ,Cloning, Molecular ,Promoter Regions, Genetic ,Gene ,Recombination, Genetic ,Mycobacterium bovis ,Sequence Analysis, DNA ,biology.organism_classification ,Virology ,Bacterial vaccine ,Expression cassette ,Plasmids - Abstract
Strain bacillus Calmette-Guerin (BCG) of Mycobacterium bovis has been used as a live bacterial vaccine to immunize more than 3 billion people against tuberculosis. In an attempt to use this vaccine strain as a vehicle for protective antigens, the gene encoding merozoite surface antigen 2 (MSA2) was amplified from strain FCC-1/HN Plasmodium falciparum genome, sequenced, and expressed in M. bovis BCG under the control of an expression cassette carrying the promoter of heat shock protein 70 (HSP70) from Mycobacterium tuberculosis. The recombinant shuttle plasmid pBCG/MSA2 was introduced into mycobacteria by electroporation, and the recombinant mycobacteria harboring pBCG/MSA2 could be induced by heating to express MSA2; the molecular mass of recombinant MSA2 was about 31 kDa. This first report of expression of the full-length P. falciparum MSA2 gene in BCG provides evidence for use of the HSP70 promoter in expressing a foreign gene in BCG and in development of BCG as a multivalent vectoral vaccine for malaria.
- Published
- 2003
- Full Text
- View/download PDF
143. Recombinant Mycobacterium bovis BCG producing the circumsporozoite protein of Plasmodium falciparum FCC-1/HN strain induces strong immune responses in BALB/c mice
- Author
-
Chunfu Zheng, Yatang Chen, and Peimei Xie
- Subjects
Plasmodium falciparum ,Protozoan Proteins ,Antibodies, Protozoan ,Booster dose ,Lymphocyte Activation ,complex mixtures ,Microbiology ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Malaria Vaccines ,parasitic diseases ,Animals ,Malaria, Falciparum ,Mice, Inbred BALB C ,Vaccines, Synthetic ,Mycobacterium bovis ,biology ,Malaria vaccine ,biology.organism_classification ,Virology ,Lymphocyte Subsets ,Circumsporozoite protein ,Infectious Diseases ,biology.protein ,Interleukin-2 ,Parasitology ,Antibody - Abstract
The current vaccine against tuberculosis, Mycobacterium bovis strain bacillus Calmette-Guerin (BCG), offers potential advantages as a live, innately immunogenic vaccine vehicle for expression and delivery of protective recombinant antigens. Malaria is one of the severest parasitic diseases in humans especially in the developing world. No efficacious vaccine is currently available. However, circumsporozoite protein (CSP) is a malaria vaccine candidate currently undergoing clinical trials. We analyzed the immune response to recombinant BCG (rBCG) vaccine expressing Plasmodium falciparum CSP (BCG-CSP) under the control of heat shock protein 70 promoter in BALB/c mice. The lymphocytes proliferative response to P. falciparum soluble antigen was significantly higher than those in the groups of BCG and normal saline, and the production of cytokines (IFN-gamma and IL-2) in response to malaria antigen was significantly higher in rBCG and BCG groups than control group of normal saline. A specific IgG antibody response against P. falciparum antigen of CSP was also characterized. The booster injection could enhance the production of cytokine, proliferation responses of spleen lymphocytes and the antibodies titer of BCG-CSP. The results in the study demonstrated that rBCG vaccine producing CSP is an appropriate vaccine for further evaluation in non-human primates.
- Published
- 2002
- Full Text
- View/download PDF
144. Herpes simplex virus 1 counteracts viperin via its virion host shutoff protein UL41
- Author
-
Shuai Wang, Mingsheng Cai, Meili Li, Kezhen Wang, Chunfu Zheng, and Guanghui Shen
- Subjects
Oxidoreductases Acting on CH-CH Group Donors ,viruses ,Immunology ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Virus Replication ,Microbiology ,Viral Proteins ,Interferon ,Virology ,medicine ,Humans ,Messenger RNA ,Host (biology) ,HEK 293 cells ,Proteins ,Virus-Cell Interactions ,Herpes simplex virus ,HEK293 Cells ,Viral replication ,Insect Science ,Viperin ,Molecular mechanism ,medicine.drug - Abstract
The interferon (IFN)-inducible viperin protein restricts a broad range of viruses. However, whether viperin plays a role during herpes simplex virus 1 (HSV-1) infection is poorly understood. In the present study, it was shown for the first time that wild-type (WT) HSV-1 infection couldn't induce viperin production, and ectopically expressed viperin inhibited the replication of UL41-null HSV-1 but not WT viruses. The underlying molecular mechanism is that UL41 counteracts viperin's antiviral activity by reducing its mRNA accumulation.
- Published
- 2014
145. Immune response induced by recombinant BCG expressing merozoite surface antigen 2 from Plasmodium falciparum
- Author
-
Chunfu Zheng, Peimei Xie, and Yatang Chen
- Subjects
Tuberculosis ,Plasmodium falciparum ,Protozoan Proteins ,Antibodies, Protozoan ,Antigens, Protozoan ,In Vitro Techniques ,Lymphocyte Activation ,Microbiology ,Apicomplexa ,Interferon-gamma ,Mice ,Immune system ,Antigen ,Malaria Vaccines ,medicine ,Animals ,Malaria, Falciparum ,Mice, Inbred BALB C ,Vaccines, Synthetic ,Mycobacterium bovis ,General Veterinary ,General Immunology and Microbiology ,biology ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,medicine.disease ,Virology ,Bacterial vaccine ,Infectious Diseases ,Immunoglobulin G ,BCG Vaccine ,Interleukin-2 ,Molecular Medicine ,Expression cassette - Abstract
Mycobacterium bovis bacillus Calmette-Guerin (BCG) has been used as a live bacterial vaccine to immunize >3 billion people against tuberculosis. In an attempt to use this vaccinal strain as a vehicle for protective antigens, the recombinant BCG (rBCG), expressing merozoite surface antigen 2 (MSA2) from Plasmodium falciparum under the control of an expression cassette carrying the promoter of heat shock protein 70 (HSP70) from M. tuberculosis, was constructed and used to immunize BABL/c mice. The administration of rBCG producing MSA2 (BCG-MSA2) resulted in the induction of a strong humoral and cellular response directed against MSA2. These results encourage the further protection testing of BCG-MSA2 vaccines in primate models.
- Published
- 2001
- Full Text
- View/download PDF
146. Molecular Cloning and Sequencing of the Circumsporozoite Protein Gene fromPlasmodium falciparumStrain FCC-1/HN and Expression of the Gene in Mycobacteria
- Author
-
Peimei Xie, Chunfu Zheng, and Yatang Chen
- Subjects
Microbiology (medical) ,Molecular Sequence Data ,Mycobacterium smegmatis ,Plasmodium falciparum ,Protozoan Proteins ,Molecular cloning ,complex mixtures ,Microbiology ,Mycobacterium tuberculosis ,parasitic diseases ,Animals ,HSP70 Heat-Shock Proteins ,Amino Acid Sequence ,Cloning, Molecular ,Promoter Regions, Genetic ,Mycobacterium bovis ,Base Sequence ,biology ,fungi ,Sequence Analysis, DNA ,biology.organism_classification ,Virology ,Circumsporozoite protein ,Bacterial vaccine ,Electroporation ,Parasitology ,Expression cassette ,Mycobacterium - Abstract
Mycobacterium bovisbacillus Calmette-Guérin (BCG) has been used as a live bacterial vaccine to immunize more than 2 billion people against tuberculosis. In an attempt to use this vaccine strain as a vehicle for protective antigens, thePlasmodium falciparumgene from strain FCC-1/HN encoding circumsporozoite protein (CSP) was amplified from theP. falciparumgenome, sequenced, and expressed inM. bovisBCG under the control of an expression cassette carrying the promoter of heat shock protein 70 (HSP70) fromMycobacterium tuberculosis. The recombinant shuttle plasmid pBCG/CSP was introduced into mycobacteria by electroporation, and the recombinant mycobacteria harboring pBCG/CSP could be induced by heating to express CSP; the molecular mass of recombinant CSP was about 42 kDa. This report of expression of the almost-full-lengthP. falciparumCSP gene in BCG provides scientific evidence for the application of the HSP70 promoter in expressing a foreign gene in BCG and in development of BCG as a multivalent vectoral vaccine for malaria.
- Published
- 2001
- Full Text
- View/download PDF
147. ID: 113
- Author
-
Chunfu Zheng, Rongtuan Lin, Sidi Mehdi Belgnaoui, Marie-Line Goulet, Alexander Sze, and Yiliu Liu
- Subjects
Messenger RNA ,RIG-I ,viruses ,Immunology ,RNA ,Signal transducing adaptor protein ,Hematology ,Biology ,Biochemistry ,Molecular biology ,eye diseases ,Sting ,Immunology and Allergy ,IRF3 ,Autocrine signalling ,Molecular Biology ,Transcription factor - Abstract
RIG-I is essential for recognizing RNA viruses with a 5’ triphosphate (ppp) signature in the cytoplasm. Upon viral RNA recognition, RIG-I recruits adaptor protein MAVS to trigger the activation of IRF3 and NF-kB transcription factors, leading to activation of antiviral response. STING has been identified as an RIG-I signaling cofactor and a critical adaptor protein required for cytosolic DNA and cyclic dinucleotides (CDNs) triggered immune responses. We have identified STING among a plethora of differentially expressed genes induced by the 5’ppp RNA; in the present study, we further detail the mechanism of STING regulation. Our data shows that virus infection induces STING expression at both the mRNA and protein levels. Furthermore STING induction is shown to be dependent on functional RIG-I signaling. STING induction by the RIG-I agonist 5’ppp RNA was recognized as a delayed event resulting from an autocrine/paracrine mechanism. Indeed, co-treatment with TNF α and IFN α has a synergistic effect on the regulation of STING expression. Depletion of STAT1 and RELA, either individually or combined, significantly diminished virus-induced STING protein expression compared with the cells. In A549 cells, the expression of interferon-response/inflammatory response genes was not affected in the absence of STING at early time points, but was significantly reduced at late time points. Furthermore, our results also unveiled an essential contribution of STING in the establishment of the 5’pppRNA induced antiviral responses during HSV1 infection. Taken together, these observations demonstrate that the STING is induced via RIG-I signaling and up-regulated STING is essential for 5’ppp RNA mediated HSV restriction.
- Published
- 2015
- Full Text
- View/download PDF
148. Herpes simplex virus 1 E3 ubiquitin ligase ICP0 protein inhibits tumor necrosis factor alpha-induced NF-κB activation by interacting with p65/RelA and p50/NF-κB1
- Author
-
Chunfu Zheng, Shuai Wang, Kezhen Wang, and Jie Zhang
- Subjects
Transcriptional Activation ,viruses ,Ubiquitin-Protein Ligases ,Immunology ,Transcription Factor RelA ,Herpesvirus 1, Human ,Microbiology ,Immediate early protein ,Immediate-Early Proteins ,Rel homology domain ,DDB1 ,Virology ,Humans ,Cell Nucleus ,biology ,Tumor Necrosis Factor-alpha ,NF-kappa B ,NF-kappa B p50 Subunit ,Herpes Simplex ,biochemical phenomena, metabolism, and nutrition ,NFKB1 ,Molecular biology ,Ubiquitin ligase ,Protein Structure, Tertiary ,Protein Transport ,HEK293 Cells ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,Tumor necrosis factor alpha ,Protein Binding - Abstract
NF-κB plays central roles in regulation of diverse biological processes, including innate and adaptive immunity and inflammation. HSV-1 is the archetypal member of the alphaherpesviruses, with a large genome encoding over 80 viral proteins, many of which are involved in virus-host interactions and show immune modulatory capabilities. In this study, we demonstrated that the HSV-1 ICP0 protein, a viral E3 ubiquitin ligase, was shown to significantly suppress tumor necrosis factor alpha (TNF-α)-mediated NF-κB activation. ICP0 was demonstrated to bind to the NF-κB subunits p65 and p50 by coimmunoprecipitation analysis. ICP0 bound to the Rel homology domain (RHD) of p65. Fluorescence microscopy demonstrated that ICP0 abolished nuclear translocation of p65 upon TNF-α stimulation. Also, ICP0 degraded p50 via its E3 ubiquitin ligase activity. The RING finger (RF) domain mutant ICP0 (ICP0-RF) lost its ability to inhibit TNF-α-mediated NF-κB activation and p65 nuclear translocation and degrade p50. Notably, the RF domain of ICP0 was sufficient to interact with p50 and abolish NF-κB reporter gene activity. Here, it is for the first time shown that HSV-1 ICP0 interacts with p65 and p50, degrades p50 through the ubiquitin-proteasome pathway, and prevents NF-κB-dependent gene expression, which may contribute to immune evasion and pathogenesis of HSV-1.
- Published
- 2013
149. Interspecies heterokaryon assay to characterize the nucleocytoplasmic shuttling of herpesviral proteins
- Author
-
Shuai, Wang, Kezheng, Wang, and Chunfu, Zheng
- Subjects
Cell Fusion ,Cell Nucleus ,Cytoplasm ,Mice ,Protein Transport ,Viral Proteins ,COS Cells ,Chlorocebus aethiops ,NIH 3T3 Cells ,Animals ,Humans ,Capsid Proteins ,Herpesvirus 1, Human - Abstract
Nucleocytoplasmic trafficking of proteins plays important roles in processes of the viral life cycle. Interspecies heterokaryon assay is one of the most effective methods to investigate the nucleocytoplasmic trafficking properties of a protein. In our lab, the interspecies heterokaryon assay has been applied to identify a few herpesviral proteins with nucleocytoplasmic shuttling property. In this chapter, the detailed information and methods of the heterokaryon assay are presented.
- Published
- 2013
150. Herpes simplex virus 1-encoded tegument protein VP16 abrogates the production of beta interferon (IFN) by inhibiting NF-κB activation and blocking IFN regulatory factor 3 to recruit its coactivator CBP
- Author
-
Liwen Ni, Shuai Wang, Chunfu Zheng, Kezhen Wang, Junji Xing, and Rongtuan Lin
- Subjects
Transcriptional Activation ,viruses ,Interferon Regulatory Factor-7 ,Immunology ,Herpesvirus 1, Human ,medicine.disease_cause ,Microbiology ,Sendai virus ,Transactivation ,Interferon ,Virology ,Coactivator ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,CREB-binding protein ,Promoter Regions, Genetic ,Vero Cells ,Innate immune system ,biology ,Transcription Factor RelA ,Herpes Simplex Virus Protein Vmw65 ,Interferon-beta ,biology.organism_classification ,CREB-Binding Protein ,Immunity, Innate ,Cell biology ,Protein Structure, Tertiary ,Herpes simplex virus ,HEK293 Cells ,Insect Science ,Host-Pathogen Interactions ,biology.protein ,Pathogenesis and Immunity ,Interferon Regulatory Factor-3 ,Signal transduction ,medicine.drug ,HeLa Cells - Abstract
Host cells activate innate immune signaling pathways to defend against invading pathogens. To survive within an infected host, viruses have evolved intricate strategies to counteract host immune responses. Herpesviruses, including herpes simplex virus type 1 (HSV-1), have large genomes and therefore have the capacity to encode numerous proteins that modulate host innate immune responses. Here we define the contribution of HSV-1 tegument protein VP16 in the inhibition of beta interferon (IFN-β) production. VP16 was demonstrated to significantly inhibit Sendai virus (SeV)-induced IFN-β production, and its transcriptional activation domain was not responsible for this inhibition activity. Additionally, VP16 blocked the activation of the NF-κB promoter induced by SeV or tumor necrosis factor alpha treatment and expression of NF-κB-dependent genes through interaction with p65. Coexpression analysis revealed that VP16 selectively blocked IFN regulatory factor 3 (IRF-3)-mediated but not IRF-7-mediated transactivation. VP16 was able to bind to IRF-3 but not IRF-7 in vivo , based on coimmunoprecipitation analysis, but it did not affect IRF-3 dimerization, nuclear translocation, or DNA binding activity. Rather, VP16 interacted with the CREB binding protein (CBP) coactivator and efficiently inhibited the formation of the transcriptional complexes IRF-3–CBP in the context of HSV-1 infection. These results illustrate that VP16 is able to block the production of IFN-β by inhibiting NF-κB activation and interfering with IRF-3 to recruit its coactivator CBP, which may be important to the early events leading to HSV-1 infection.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.