Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Microbiol Resour Announc ; 13(4): e0124623, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38451104

ABSTRACT

Bacillus halotolerans F29-3, a Gram-positive bacterium, is recognized for its synthesis of the antifungal substance fengycin. This announcement introduces the complete genome sequence and provides insights into the genetic products related to antibiotic secondary metabolites, including non-ribosomal peptide synthetase (NRPS), polyketide synthase (PKS), and NRPS/PKS combination.

3.
J Microbiol Immunol Infect ; 50(6): 755-762, 2017 Dec.
Article in English | MEDLINE | ID: mdl-26857334

ABSTRACT

BACKGROUND: Fengycin is a lipopeptide antibiotic synthesized nonribosomally by five fengycin synthetases. These enzymes are linked in a specific order to form the complex. This study investigates how these enzymes interact in the complex and analyzes the regions in the enzymes that are critical to the interactions. METHODS: Deletions were generated in the fengycin synthetases. The interaction of these mutant proteins with their partner enzymes in the complex was analyzed in vitro by a glutathione S-transferase (GST) or nickel pulldown assay. RESULTS: The communication-mediating donor (COM-D) domains of the fengycin synthetases, when fused to GST, specifically pulled down their downstream partner enzymes in the GST-pulldown assays. The communication-mediating acceptor (COM-A) domains were required for binding between two partner enzymes, although the domains alone did not confer specificity of the binding to their upstream partner enzymes. This study found that the COM-A domain, the condensation domain, and a portion of the adenylation domain in fengycin synthetase B (FenB) were required for specific binding to fengycin synthetase A (FenA). CONCLUSION: The interaction between the COM-D and COM-A domains in two partner enzymes is critical for nonribosomal peptide synthesis. The COM-A domain alone is insufficient for interacting with its upstream partner enzyme in the enzyme complex with specificity; a region that contains COM-A, condensation, and a portion of adenylation domains in the downstream partner enzyme is required.


Subject(s)
Bacillus subtilis/metabolism , Lipopeptides/biosynthesis , Peptide Biosynthesis, Nucleic Acid-Independent/physiology , Peptide Synthases/genetics , Peptide Synthases/metabolism , Protein Interaction Mapping , Amino Acid Sequence , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Escherichia coli/metabolism , Glutathione Transferase/metabolism , Protein Binding , Protein Domains/genetics , Sequence Deletion/genetics
4.
PLoS Pathog ; 12(10): e1005918, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27698494

ABSTRACT

The switch between latency and the lytic cycle of Kaposi's sarcoma-associated herpesvirus (KSHV) is controlled by the expression of virally encoded ORF50 protein. Thus far, the regulatory mechanism underlying the protein stability of ORF50 is unknown. Our earlier studies have demonstrated that a protein abundance regulatory signal (PARS) at the ORF50 C-terminal region modulates its protein abundance. The PARS region consists of PARS-I (aa 490-535) and PARS-II (aa 590-650), and mutations in either component result in abundant expression of ORF50. Here, we show that ORF50 protein is polyubiquitinated and its abundance is controlled through the proteasomal degradation pathway. The PARS-I motif mainly functions as a nuclear localization signal in the control of ORF50 abundance, whereas the PARS-II motif is required for the binding of ubiquitin enzymes in the nucleus. We find that human oncoprotein MDM2, an ubiquitin E3 ligase, is capable of interacting with ORF50 and promoting ORF50 degradation in cells. The interaction domains between both proteins are mapped to the PARS region of ORF50 and the N-terminal 220-aa region of MDM2. Additionally, we identify lysine residues at positions 152 and 154 in the N-terminal domain of ORF50 critically involved in MDM2-mediated downregulation of ORF50 levels. Within KSHV-infected cells, the levels of MDM2 were greatly reduced during viral lytic cycle and genetic knockdown of MDM2 in these cells favored the enhancement of ORF50 expression, supporting that MDM2 is a negative regulator of ORF50 expression. Collectively, the study elucidates the regulatory mechanism of ORF50 stability and implicates that MDM2 may have a significant role in the maintenance of viral latency by lowering basal level of ORF50.


Subject(s)
Gene Expression Regulation, Viral/physiology , Herpesviridae Infections/metabolism , Immediate-Early Proteins/biosynthesis , Proto-Oncogene Proteins c-mdm2/metabolism , Trans-Activators/biosynthesis , Virus Latency/physiology , Cell Line , Fluorescent Antibody Technique , Herpesvirus 8, Human , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Confocal , Protein Stability
5.
PLoS Pathog ; 12(10): e1005959, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27780225

ABSTRACT

The 5' untranslated region (5' UTR) of the enterovirus 71 (EV71) RNA genome contains an internal ribosome entry site (IRES) that is indispensable for viral protein translation. Due to the limited coding capacity of their RNA genomes, EV71 and other picornaviruses typically recruit host factors, known as IRES trans-acting factors (ITAFs), to mediate IRES-dependent translation. Here, we show that EV71 viral proteinase 2A is capable of cleaving far upstream element-binding protein 1 (FBP1), a positive ITAF that directly binds to the EV71 5' UTR linker region to promote viral IRES-driven translation. The cleavage occurs at the Gly-371 residue of FBP1 during the EV71 infection process, and this generates a functional cleavage product, FBP11-371. Interestingly, the cleavage product acts to promote viral IRES activity. Footprinting analysis and gel mobility shift assay results showed that FBP11-371 similarly binds to the EV71 5' UTR linker region, but at a different site from full-length FBP1; moreover, FBP1 and FBP11-371 were found to act additively to promote IRES-mediated translation and virus yield. Our findings expand the current understanding of virus-host interactions with regard to viral recruitment and modulation of ITAFs, and provide new insights into translational control during viral infection.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Enterovirus A, Human , Gene Expression Regulation, Viral/physiology , Host-Parasite Interactions/physiology , Internal Ribosome Entry Sites/physiology , Viral Proteins/metabolism , 5' Untranslated Regions/physiology , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Humans , Immunoblotting , Immunoprecipitation , Internal Ribosome Entry Sites/genetics , Protein Biosynthesis/physiology , RNA-Binding Proteins
6.
J Cell Mol Med ; 20(7): 1295-306, 2016 07.
Article in English | MEDLINE | ID: mdl-26969027

ABSTRACT

Cullin 4A (Cul4A) has been observed to be overexpressed in various cancers. In this study, the role of Cul4A in the growth and chemosensitivity in lung cancer cells were studied. We showed that Cul4A is overexpressed in lung cancer cells and tissues. Knockdown of the Cul4A expression by shRNA in lung cancer cells resulted in decreased cellular proliferation and growth in lung cancer cells. Increased sensitivity to gemcitabine, a chemotherapy drug, was also noted in those Cul4A knockdown lung cancer cells. Moreover, increased expression of p21, transforming growth factor (TGF)-ß inducible early gene-1 (TIEG1) and TGF beta-induced (TGFBI) was observed in lung cancer cells after Cul4A knockdown, which may be partially related to increased chemosensitivity to gemcitabine. G0/G1 cell cycle arrest was also noted after Cul4A knockdown. Notably, decreased tumour growth and increased chemosensitivity to gemcitabine were also noted after Cul4A knockdown in lung cancer xenograft nude mice models. In summary, our study showed that targeting Cul4A with RNAi or other techniques may provide a possible insight to the development of lung cancer therapy in the future.


Subject(s)
Antineoplastic Agents/pharmacology , Cullin Proteins/metabolism , Gene Knockdown Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Humans , Inhibitory Concentration 50 , Mice, Inbred BALB C , Neoplasm Proteins/metabolism , RNA, Small Interfering/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
7.
Front Microbiol ; 7: 2129, 2016.
Article in English | MEDLINE | ID: mdl-28105027

ABSTRACT

Replication and transcription activator (Rta), a key protein expressed by Epstein-Barr virus (EBV) during the immediate-early stage of the lytic cycle, is responsible for the activation of viral lytic genes. In this study, GST-pulldown and coimmunoprecipitation assays showed that Rta interacts in vitro and in vivo with TRIM5α, a host factor known to be involved in the restriction of retroviral infections. Confocal microscopy results revealed that Rta colocalizes with TRIM5α in the nucleus during lytic progression. The interaction involves 190 amino acids in the N-terminal of Rta and the RING domain in TRIM5α, and it was further found that TRIM5α acts as an E3 ubiquitin ligase to promote Rta ubiquitination. Overexpression of TRIM5α reduced the transactivating capabilities of Rta, while reducing TRIM5α expression enhanced EBV lytic protein expression and DNA replication. Taken together, these results point to a critical role for TRIM5α in attenuating EBV lytic progression through the targeting of Rta for ubiquitination, and suggest that the restrictive capabilities of TRIM5α may go beyond retroviral infections.

8.
PLoS One ; 10(12): e0144306, 2015.
Article in English | MEDLINE | ID: mdl-26669836

ABSTRACT

The superior antimicrobial properties of silver nanoparticles (Ag NPs) are well-documented, but the exact mechanisms underlying Ag-NP microbial toxicity remain the subject of intense debate. Here, we show that Ag-NP concentrations as low as 10 ppm exert significant toxicity against Bacillus subtilis, a beneficial bacterium ubiquitous in the soil. Growth arrest and chromosomal DNA degradation were observed, and flow cytometric quantification of propidium iodide (PI) staining also revealed that Ag-NP concentrations of 25 ppm and above increased membrane permeability. RedoxSensor content analysis and Phag-GFP expression analysis further indicated that reductase activity and cytosolic protein expression decreased in B. subtilis cells treated with 10-50 ppm of Ag NPs. We conducted X-ray absorption near-edge structure (XANES) and extended X-ray absorption fine structure (EXAFS) analyses to directly clarify the valence and fine structure of Ag atoms in B. subtilis cells placed in contact with Ag NPs. The results confirmed the Ag species in Ag NP-treated B. subtilis cells as Ag2O, indicating that Ag-NP toxicity is likely mediated by released Ag+ ions from Ag NPs, which penetrate bacterial cells and are subsequently oxidized intracellularly to Ag2O. These findings provide conclusive evidence for the role of Ag+ ions in Ag-NP microbial toxicity, and suggest that the impact of inappropriately disposed Ag NPs to soil and water ecosystems may warrant further investigation.


Subject(s)
Anti-Infective Agents/pharmacology , Bacillus subtilis/drug effects , Metal Nanoparticles/chemistry , Silver/pharmacology , Bacillus subtilis/growth & development , Chromosomes, Bacterial/metabolism , Crystallography, X-Ray , DNA, Bacterial/metabolism , Green Fluorescent Proteins/metabolism , Ions , Microbial Sensitivity Tests , Oxidation-Reduction , Particle Size , Reference Standards , Staining and Labeling , X-Ray Absorption Spectroscopy
9.
Front Microbiol ; 6: 1017, 2015.
Article in English | MEDLINE | ID: mdl-26557106

ABSTRACT

Many Bacillus subtilis strains swarm, often forming colonies with tendrils on agar medium. It is known that B. subtilis swarming requires flagella and a biosurfactant, surfactin. In this study, we find that water surface tension plays a role in swarming dynamics. B. subtilis colonies were found to contain water, and when a low amount of surfactin is produced, the water surface tension of the colony restricts expansion, causing bacterial density to rise. The increased density induces a quorum sensing response that leads to heightened production of surfactin, which then weakens water surface tension to allow colony expansion. When the barrier formed by water surface tension is breached at a specific location, a stream of bacteria swarms out of the colony to form a tendril. If a B. subtilis strain produces surfactin at levels that can substantially weaken the overall water surface tension of the colony, water floods the agar surface in a thin layer, within which bacteria swarm and migrate rapidly. This study sheds light on the role of water surface tension in regulating B. subtilis swarming, and provides insight into the mechanisms underlying swarming initiation and tendril formation.

10.
J Gen Virol ; 96(9): 2855-2866, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26297580

ABSTRACT

During its lytic cycle, Epstein-Barr virus (EBV) expresses Rta, a factor encoded by BRLF1 that activates the transcription of viral lytic genes. We found that upstream stimulating factor (USF) binds to E1, one of the five E boxes located at - 79 in the BRLF1 promoter (Rp), to activate BRLF1 transcription. Furthermore, Rta was shown to interact with USF1 in coimmunoprecipitation and glutathione S-transferase (GST)-pulldown assays, and confocal laser-scanning microscopy further confirmed that these two proteins colocalize in the nucleus. Rta was also found to bind with the E1 sequence in a biotin-labelled E1 probe, but only in the presence of USF1, suggesting that these two proteins likely form a complex on E1. We subsequently constructed p188mSZ, a reporter plasmid that contained the sequence from - 188 to +5 in Rp, within which the Sp1 site and Zta response element were mutated. In EBV-negative Akata cells cotransfected with p188mSZ and plasmids expressing USF1 and Rta, synergistic activation of Rp transcription was observed. However, after mutating the E1 sequence in p188mSZ, USF1 and Rta were no longer able to transactivate Rp, indicating that Rta autoregulates BRLF1 transcription via its interaction with USF1 on E1. This study showed that pUSF1 transfection after EBV lytic induction in P3HR1 cells increases Rta expression, indicating that USF1 activates Rta expression after the virus enters the lytic cycle. Together, these results reveal a novel mechanism by which USF interacts with Rta to promote viral lytic development, and provide additional insight into the viral-host interactions of EBV.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/genetics , Immediate-Early Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Activation , Upstream Stimulatory Factors/metabolism , Base Sequence , Binding Sites , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/metabolism , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Trans-Activators/chemistry , Upstream Stimulatory Factors/genetics
11.
J Virol ; 89(17): 8922-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26085145

ABSTRACT

The Epstein-Barr virus (EBV) capsid contains a major capsid protein, VCA; two minor capsid proteins, BDLF1 and BORF1; and a small capsid protein, BFRF3. During the lytic cycle, these capsid proteins are synthesized and imported into the host nucleus for capsid assembly. This study finds that EBV capsid proteins colocalize with promyelocytic leukemia (PML) nuclear bodies (NBs) in P3HR1 cells during the viral lytic cycle, appearing as nuclear speckles under a confocal laser scanning microscope. In a glutathione S-transferase pulldown study, we show that BORF1 interacts with PML-NBs in vitro. BORF1 also colocalizes with PML-NBs in EBV-negative Akata cells after transfection and is responsible for bringing VCA and the VCA-BFRF3 complex from the cytoplasm to PML-NBs in the nucleus. Furthermore, BDLF1 is dispersed throughout the cell when expressed alone but colocalizes with PML-NBs when BORF1 is also present in the cell. In addition, this study finds that knockdown of PML expression by short hairpin RNA does not influence the intracellular levels of capsid proteins but reduces the number of viral particles produced by P3HR1 cells. Together, these results demonstrate that BORF1 plays a critical role in bringing capsid proteins to PML-NBs, which may likely be the assembly sites of EBV capsids. The mechanisms elucidated in this study are critical to understanding the process of EBV capsid assembly. IMPORTANCE Capsid assembly is an important event during the Epstein-Barr virus (EBV) lytic cycle, as this process is required for the production of virions. In this study, confocal microscopy revealed that the EBV capsid protein BORF1 interacts with promyelocytic leukemia (PML) nuclear bodies (NBs) in the host nucleus and is responsible for transporting the other EBV capsid proteins, including VCA, BDLF1, and BFRF3, to these subnuclear locations prior to initiation of capsid assembly. This study also found that knockdown of PML expression by short hairpin RNA significantly reduces EBV capsid assembly capabilities. This enhanced understanding of capsid assembly offers potential for the development of novel antiviral strategies and therapies that can prevent the propagation and spread of EBV.


Subject(s)
Active Transport, Cell Nucleus/genetics , Antigens, Viral/metabolism , Capsid Proteins/metabolism , Capsid/metabolism , Herpesvirus 4, Human/metabolism , Neoplasm Proteins/metabolism , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Capsid Proteins/biosynthesis , Capsid Proteins/genetics , Cell Line, Tumor , HEK293 Cells , Herpesvirus 4, Human/genetics , Humans , Leukemia, Promyelocytic, Acute/virology , Microscopy, Confocal , Nuclear Proteins/metabolism , Protein Transport/genetics , RNA Interference , RNA, Small Interfering
12.
PLoS One ; 10(4): e0124216, 2015.
Article in English | MEDLINE | ID: mdl-25876106

ABSTRACT

Staphylococcus aureus is an important pathogen that forms biofilms on the surfaces of medical implants. Biofilm formation by S. aureus is associated with the production of poly N-acetylglucosamine (PNAG), also referred to as polysaccharide intercellular adhesin (PIA), which mediates bacterial adhesion, leading to the accumulation of bacteria on solid surfaces. This study shows that the ability of S. aureus SA113 to adhere to nasal epithelial cells is reduced after the deletion of the ica operon, which contains genes encoding PIA/PNAG synthesis. However, this ability is restored after a plasmid carrying the entire ica operon is transformed into the mutant strain, S. aureus SA113Δica, showing that the synthesis of PIA/PNAG is important for adhesion to epithelial cells. Additionally, S. carnosus TM300, which does not produce PIA/PNAG, forms a biofilm and adheres to epithelial cells after the bacteria are transformed with a PIA/PNAG-expressing plasmid, pTXicaADBC. The adhesion of S. carnosus TM300 to epithelial cells is also demonstrated by adding purified exopolysaccharide (EPS), which contains PIA/PNAG, to the bacteria. In addition, using a mouse model, we find that the abscess lesions and bacterial burden in lung tissues is higher in mice infected with S. aureus SA113 than in those infected with the mutant strain, S. aureus SA113Δica. The results indicate that PIA/PNAG promotes the adhesion of S. aureus to human nasal epithelial cells and lung infections in a mouse model. This study elucidates a mechanism that is important to the pathogenesis of S. aureus infections.


Subject(s)
Bacterial Adhesion/genetics , Biofilms/growth & development , Gene Expression Regulation, Bacterial , Polysaccharides, Bacterial/metabolism , Staphylococcal Infections/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Animals , Bacterial Load/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cell Line, Tumor , Disease Models, Animal , Epithelial Cells/microbiology , Epithelial Cells/pathology , Genetic Complementation Test , Humans , Lung/microbiology , Lung/pathology , Mice , Molecular Sequence Data , Operon , Plasmids/chemistry , Plasmids/metabolism , Sequence Deletion , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Transformation, Bacterial , beta-Glucans/metabolism
13.
J Gen Virol ; 96(8): 2336-2348, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25900136

ABSTRACT

Epstein-Barr virus (EBV) expresses two immediate-early proteins, Rta and Zta, which are key transcription factors that can form a complex with MCAF1 at Zta-responsive elements (ZREs) to synergistically activate several viral lytic genes. Our previous research indicated that RanBPM interacts with Rta and enhances Rta sumoylation. Here we showed that RanBPM binds to Zta in vitro and in vivo, and acts as an intermediary protein in Rta-Zta complex formation. The Rta-RanBPM-Zta complex was observed to bind with ZREs in the transcriptional activation of key viral genes, such as BHLF1 and BHRF1, while the introduction of RanBPM short hairpin RNA (shRNA) subsequently reduced the synergistic activity of Zta and Rta. RanBPM was found to enhance Zta-dependent transcriptional activity via the inhibition of Zta sumoylation. Interestingly, Z-K12R, a sumoylation-defective mutant of Zta, demonstrated transcriptional activation capabilities that were stronger than those of Zta and apparently unaffected by RanBPM modulation. Finally, RanBPM silencing inhibited the expression of lytic proteins. Taken together, these results shed light on the mechanisms by which RanBPM regulates Zta-mediated transcriptional activation, and point to an important role for RanBPM in EBV lytic progression.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Nuclear Proteins/genetics , Protein Binding , Trans-Activators/genetics
14.
J Virol ; 88(20): 12133-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25122800

ABSTRACT

Autophagy is an intracellular degradation pathway that provides a host defense mechanism against intracellular pathogens. However, many viruses exploit this mechanism to promote their replication. This study shows that lytic induction of Epstein-Barr virus (EBV) increases the membrane-bound form of LC3 (LC3-II) and LC3-containing punctate structures in EBV-positive cells. Transfecting 293T cells with a plasmid that expresses Rta also induces autophagy, revealing that Rta is responsible for autophagic activation. The activation involves Atg5, a key component of autophagy, but not the mTOR pathway. The expression of Rta also activates the transcription of the genes that participate in the formation of autophagosomes, including LC3A, LC3B, and ATG9B genes, as well as those that are involved in the regulation of autophagy, including the genes TNF, IRGM, and TRAIL. Additionally, treatment with U0126 inhibits the Rta-induced autophagy and the expression of autophagy genes, indicating that the autophagic activation is caused by the activation of extracellular signal-regulated kinase (ERK) signaling by Rta. Finally, the inhibition of autophagic activity by an autophagy inhibitor, 3-methyladenine, or Atg5 small interfering RNA, reduces the expression of EBV lytic proteins and the production of viral particles, revealing that autophagy is critical to EBV lytic progression. This investigation reveals how an EBV-encoded transcription factor promotes autophagy to affect viral lytic development.


Subject(s)
Autophagy , Extracellular Signal-Regulated MAP Kinases/metabolism , Herpesvirus 4, Human/immunology , Immediate-Early Proteins/physiology , Trans-Activators/physiology , Base Sequence , DNA Primers , HEK293 Cells , Humans , Microscopy, Electron, Transmission , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
15.
PLoS One ; 9(3): e90698, 2014.
Article in English | MEDLINE | ID: mdl-24598729

ABSTRACT

Epstein-Barr virus (EBV) expresses two transcription factors, Rta and Zta, which are involved in the transcriptional activation of EBV lytic genes. This study sought to elucidate the mechanism by which Rta activates transcription of the Zta-encoding gene, BZLF1, through the ZII element in the gene promoter. In a DNA affinity precipitation assay, ATF2 was found to associate with an Rta-interacting protein, MCAF1, at the ZII element. The interaction between Rta, MCAF1, and ATF2 at the same site in the ZII region was further verified in vivo by chromatin immunoprecipitation assay. The complex appears to be crucial for the activation of BZLF1 transcription, as the overexpression of two ATF2-dominant negative mutants, or the introduction of MCAF1 siRNA into 293T cells, were both found to substantially reduce Rta-mediated transcription levels of BZLF1. Moreover, this study also found that the Rta-MCAF1-ATF2 complex binds to a typical AP-1 binding sequence on the promoter of BMRF2, a key viral gene for EBV infection. Mutation of this sequence decreased Rta-mediated promoter activity significantly. Taken together, these results indicate a critical role for MCAF1 in AP-1-dependent Rta activation of BZLF1 transcription.


Subject(s)
Herpesvirus 4, Human/genetics , Trans-Activators/genetics , Transcription Factors/metabolism , Transcription, Genetic , Activating Transcription Factor 2/chemistry , Activating Transcription Factor 2/metabolism , Binding Sites , Cell Line, Tumor , Humans , Immunoprecipitation , Membrane Glycoproteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Repressor Proteins , Transcription Factor AP-1/metabolism , Transcription Factors/chemistry , Transcriptional Activation/genetics , Viral Proteins/genetics
16.
PLoS One ; 8(4): e61668, 2013.
Article in English | MEDLINE | ID: mdl-23613898

ABSTRACT

Plasmid pSW200 from Pantoea stewartii contains 41 copies of 15-bp repeats and has a replicon that is homologous to that of ColE1. Although deleting the repeats (pSW207) does not change the copy number and stability of the plasmid. The plasmid becomes unstable and is rapidly lost from the host when a homoplasmid with the repeats (pSW201) is present. Deleting the repeats is found to reduce the transcriptional activity of RNAIp and RNAIIp by about 30%, indicating that the repeats promote the transcription of RNAI and RNAII, and how the RNAI that is synthesized by pSW201 inhibits the replication of pSW207. The immunoblot analysis herein demonstrates that RNA polymerase ß subunit and σ(70) in the lysate from Escherichia coli MG1655 bind to a biotin-labeled DNA probe that contains the entire sequence of the repeat region. Electrophoretic mobility shift assay also reveals that purified RNA polymerase shifts a DNA probe that contains four copies of the repeats. These results thus obtained reveal that RNA polymerase holoenzyme binds to the repeats. The repeats also exchange RNA polymerase with RNAIp and RNAIIp in vitro, revealing the mechanism by which the transcription is promoted. This investigation elucidates a mechanism by which a plasmid prevents the invasion of an incompatible plasmid and maintains its stability in the host cell during evolution.


Subject(s)
Plasmids/genetics , Repetitive Sequences, Nucleic Acid/genetics , DNA-Directed RNA Polymerases/metabolism , Electrophoretic Mobility Shift Assay , Pantoea/genetics
17.
J Virol ; 86(18): 9647-55, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22740416

ABSTRACT

Epstein-Barr virus (EBV) BBLF1 shares 13 to 15% amino acid sequence identities with the herpes simplex virus 1 UL11 and cytomegalovirus UL99 tegument proteins, which are involved in the final envelopment during viral maturation. This study demonstrates that BBLF1 is a myristoylated and palmitoylated protein, as are UL11 and UL99. Myristoylation of BBLF1 both facilitates its membrane anchoring and stabilizes it. BBLF1 is shown to localize to the trans-Golgi network (TGN) along with gp350/220, a site where final envelopment of EBV particles takes place. The localization of BBLF1 at the TGN requires myristoylation and two acidic clusters, which interact with PACS-1, a cytosolic protein, to mediate retrograde transport from the endosomes to the TGN. Knockdown of the expression of BBLF1 during EBV lytic replication reduces the production of virus particles, demonstrating the requirement of BBLF1 to achieve optimal production of virus particles. BBLF1 is hypothesized to facilitate the budding of tegumented capsid into glycoprotein-embedded membrane during viral maturation.


Subject(s)
Herpesvirus 4, Human/physiology , Viral Proteins/physiology , Amino Acid Sequence , Base Sequence , Biological Transport, Active , DNA, Viral/genetics , Gene Knockdown Techniques , HEK293 Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Lipoylation , Molecular Sequence Data , Myristic Acid/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vesicular Transport Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication , trans-Golgi Network/virology
18.
Mol Cells ; 31(6): 547-52, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21533551

ABSTRACT

MCRS2 is an oncoprotein that is sequestered in the nucleolus. When in the nucleolus, it promotes the transcription of the rRNA gene. MCRS2 also brings proteins into the nucleolus to change their function. This study analyzes the sequence of MCRS2 and determines that the nuclear localization signal, which has the sequence KRKK, is situated between amino acids 66 and 69. Meanwhile, MCRS2 contains a bipartite nucleolar localization signal, which comprises a KKSK motif, located between amino acids 133 and 136, and a downstream 152-amino acid region, from amino acid 314 to 465. The results of this study are important to understand the function of MCRS2.


Subject(s)
Cell Nucleolus/metabolism , Nuclear Localization Signals/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Microscopy, Confocal , Molecular Sequence Data , Nuclear Localization Signals/genetics , Nuclear Proteins/genetics , Protein Structure, Tertiary , Protein Transport , RNA-Binding Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion
19.
J Gen Virol ; 92(Pt 8): 1760-1768, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21490247

ABSTRACT

Epstein-Barr virus (EBV) expresses two transcription factors, Rta and Zta, during the immediate-early stage of the lytic cycle to activate the transcription of early and late genes. This study finds that 0.31 mM protoapigenone from Thelypteris torresiana (Gaud.) inhibits the expression of EBV lytic proteins, including Rta, Zta, EA-D and VCA, in P3HR1 cells after lytic induction with 12-O-tetradecanoylphorbol-13-acetate and sodium butyrate. The lack of expression of EBV lytic proteins after protoapigenone treatment is attributed to the inhibition of the transactivation function of Zta because protoapigenone reduces the transactivation activity of Zta and Gal4-Zta, which contains the transactivation domain of Zta fused with Gal4. In contrast, protoapigenone does not affect the ability of Rta to activate a promoter that contains an Rta-response element, showing that the inhibition is unrelated to Rta. Furthermore, in a lactate dehydrogenase assay, protoapigenone is not toxic to P3HR1 cells at the concentrations that inhibit the function of Zta, showing that protoapigenone is valuable for studying the function of Zta and preventing EBV lytic proliferation.


Subject(s)
Cyclohexanones/pharmacology , Down-Regulation/drug effects , Ferns/chemistry , Flavones/pharmacology , Herpesvirus 4, Human/physiology , Plant Extracts/pharmacology , Cell Line , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/genetics , Humans , Viral Proteins/genetics , Viral Proteins/metabolism
20.
J Virol ; 85(4): 1615-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21147928

ABSTRACT

The capsids of herpesviruses, which comprise major and minor capsid proteins, have a common icosahedral structure with 162 capsomers. An electron microscopic study shows that Epstein-Barr virus (EBV) capsids in the nucleus are immunolabeled by anti-BDLF1 and anti-BORF1 antibodies, indicating that BDLF1 and BORF1 are the minor capsid proteins of EBV. Cross-linking and electrophoresis studies of purified BDLF1 and BORF1 revealed that these two proteins form a triplex that is similar to that formed by the minor capsid proteins, VP19C and VP23, of herpes simplex virus type 1 (HSV-1). Although the interaction between VP23, a homolog of BDLF1, and the major capsid protein VP5 could not be verified biochemically in earlier studies, the interaction between BDLF1 and the EBV major capsid protein, viral capsid antigen (VCA), can be confirmed by glutathione S-transferase (GST) pulldown assay and coimmunoprecipitation. Additionally, in HSV-1, VP5 interacts with only the middle region of VP19C; in EBV, VCA interacts with both the N-terminal and middle regions of BORF1, a homolog of VP19C, revealing that the proteins in the EBV triplex interact with the major capsid protein differently from those in HSV-1. A GST pulldown study also identifies the oligomerization domains in VCA and the dimerization domain in BDLF1. The results presented herein reveal how the EBV capsid proteins interact and thereby improve our understanding of the capsid structure of the virus.


Subject(s)
Antigens, Viral/metabolism , Capsid Proteins/metabolism , Capsid/ultrastructure , Herpesvirus 4, Human/metabolism , Animals , Antigens, Viral/chemistry , Antigens, Viral/genetics , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Herpesvirus 1, Human/metabolism , Herpesvirus 4, Human/ultrastructure , Humans , Mice , Microscopy, Electron/methods , Protein Interaction Mapping , Rats , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/metabolism , Virus Assembly
SELECTION OF CITATIONS
SEARCH DETAIL