Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Med Virol ; 96(3): e29504, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38445794

ABSTRACT

While most NOD-like receptors (NLRs) are predominately expressed by innate immune cells, NLRC3, an inhibitory NLR of immune signaling, exhibits the highest expression in lymphocytes. The role of NLRC3 or any NLRs in B lymphocytes is completely unknown. Gammaherpesviruses, including human Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV-68), establish latent infection in B lymphocytes, which requires elevated NF-κB. This study shows that during latent EBV infection of human B cells, viral-encoded latent membrane protein 1 (LMP1) decreases NLRC3 transcript. LMP1-induced-NF-κB activation suppresses the promoter activity of NLRC3 via p65 binding to the promoter. Conversely, NLRC3 inhibits NF-κB activation by promoting the degradation of LMP1 in a proteasome-dependent manner. In vivo, MHV-68 infection reduces Nlrc3 transcripts in splenocytes, and Nlrc3-deficient mice show greater viral latency than controls. These results reveal a bidirectional regulatory circuit in B lymphocytes, where viral latent protein LMP1 reduces NLRC3 expression, while NLRC3 disrupts gammaherpesvirus latency, which is an important step for tumorigenesis.


Subject(s)
Epstein-Barr Virus Infections , Virus Latency , Animals , Humans , Mice , Herpesvirus 4, Human/genetics , NF-kappa B , B-Lymphocytes , Intercellular Signaling Peptides and Proteins
2.
Arch Pharm Res ; 46(7): 598-615, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37563335

ABSTRACT

The G-quadruplex (G4) formed in single-stranded DNAs or RNAs plays a key role in diverse biological processes and is considered as a potential antiviral target. In the genome of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), 25 putative G4-forming sequences are predicted; however, the effects of G4-binding ligands on SARS-CoV-2 replication have not been studied in the context of viral infection. In this study, we investigated whether G4-ligands suppressed SARS-CoV-2 replication and whether their antiviral activity involved stabilization of viral RNA G4s and suppression of viral gene expression. We found that pyridostatin (PDS) suppressed viral gene expression and genome replication as effectively as the RNA polymerase inhibitor remdesivir. Biophysical analyses revealed that the 25 predicted G4s in the SARS-CoV-2 genome formed a parallel G4 structure. In particular, G4-644 and G4-3467 located in the 5' region of ORF1a, formed a G4 structure that could be effectively stabilized by PDS. We also showed that PDS significantly suppressed translation of the reporter genes containing these G4s. Taken together, our results demonstrate that stabilization of RNA G4s by PDS in the SARS-CoV-2 genome inhibits viral infection via translational suppression, highlighting the therapeutic potential of G4-ligands in SARS-CoV-2 infection.


Subject(s)
COVID-19 , G-Quadruplexes , Humans , SARS-CoV-2 , Ligands , Antiviral Agents/therapeutic use
3.
PLoS Pathog ; 19(1): e1011095, 2023 01.
Article in English | MEDLINE | ID: mdl-36630443

ABSTRACT

G-quadruplex (G4) formed by repetitive guanosine-rich sequences plays important roles in diverse cellular processes; however, its roles in viral infection are not fully understood. In this study, we investigated the genome-wide distribution of G4-forming sequences (G4 motifs) in Varicella-Zoster virus (VZV) and found that G4 motifs are enriched in the internal repeat short and the terminal repeat short regions flanking the unique short region and also in some reiteration (R) sequence regions. A high density of G4 motifs in the R2 region was found on the template strand of ORF14, which encodes glycoprotein C (gC), a virulent factor for viral growth in skin. Analyses such as circular dichroism spectroscopy, thermal difference spectra, and native polyacrylamide gel electrophoresis with oligodeoxynucleotides demonstrated that several G4 motifs in ORF14 form stable G4 structures. In transfection assays, gC expression from the G4-disrupted ORF14 gene was increased at the transcriptional level and became more resistant to suppression by G4-ligand treatment. The recombinant virus containing the G4-disrupted ORF14 gene expressed a higher level of gC mRNA, while it showed a slightly reduced growth. This G4-disrupted ORF14 virus produced smaller plaques than the wild-type virus. Our results demonstrate that G4 formation via reiteration sequences suppresses gC expression during VZV infection and regulates viral cell-to-cell spread.


Subject(s)
G-Quadruplexes , Herpesvirus 3, Human/genetics , Viral Envelope Proteins/genetics , Genome , Circular Dichroism
4.
J Cell Biol ; 222(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36542408

ABSTRACT

Liquid-liquid phase separation (LLPS) has emerged as a fundamental mechanism to compartmentalize biomolecules into membraneless organelles. In this issue, Zhou et al. (2022. J. Cell Biol.https://doi.org/10.1083/jcb.202201088), report that MHV-68 ORF52 undergoes LLPS to form cytoplasmic virion assembly compartments, regulating the spatiotemporal compartmentalization of viral components.


Subject(s)
Cytoplasm , Herpesviridae , Virus Assembly , Cytoplasm/virology , Herpesviridae/physiology , Organelles
5.
J Virol ; 96(21): e0037122, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36286483

ABSTRACT

Gammaherpesviruses, including Epstein-Barr virus (EBV), are important human pathogens because they are associated with various tumors. Poly(ADP-ribose) polymerase 1 (PARP1) is a multifunctional host nuclear protein responsible for poly(ADP-ribosyl)ation (PARylation) of target proteins. While PARP1 acts as a negative regulator that suppresses the lytic replication of gammaherpesviruses, viruses are often equipped with various strategies to overcome PARP1 inhibition. However, the mechanisms of how EBV may modulate a repressive host protein, PARP1, are still elusive. In this study, we found that EBV reactivation induced PARP1 downregulation in EBV-infected cells. EBV DNA polymerase processivity factor EA-D, encoded by the BMRF1 gene, directly interacted with the central automodification domain (AD) of PARP1 and was necessary and sufficient to downregulate PARP1 via K29-linked polyubiquitination. Moreover, knockdown of EA-D in B95.8 cells restored PARP1 levels and abrogated the expression of ZTA (also known as ZEBRA), a switch molecule of the EBV life cycle during reactivation. Interestingly, PARP1 PARylated RTA, another key switch molecule, and decreased RTA transactivation on the promoters of the ZTA, BMRF1, and BMLF1 genes. EA-D alleviated the PARylation of RTA and further enhanced RTA-mediated transactivation of these lytic promoters in reporter assays. Taken together, our results suggest that EBV viral processivity factor plays a key role in facilitating lytic replication by inducing PARP1 degradation via its interaction with the PARP1 AD, which is a highly conserved mechanism among gammaherpesviruses to counteract host repressive activity of PARP1 against viral lytic replication. IMPORTANCE PARP1 acts as a negative regulator of lytic replication in EBV. To successfully enter the reactivation cycle, EBV has developed multiple strategies to counteract the host's repressive mechanisms. In this study, we investigated how EBV manipulated the host repressive factor PARP1 to facilitate lytic replication. The EBV processivity factor EA-D downregulated PARP1 in a proteasome-dependent manner via its direct binding with PARP1 AD. The knockdown of EA-D restored the PARP1 level and inhibited ZTA expression during reactivation. Interestingly, PARP1 PARylated RTA and EA-D reduced the PARylation of RTA, thereby promoting the ZTA promoter activity. These results suggest that EA-D plays a key role in EBV lytic replication by inducing PARP1 degradation in addition to supporting DNA replication as a viral processivity factor. Given that the KSHV processivity factor also induces PARP1 degradation and enhances RTA function, gammaherpesviruses share a conserved molecular mechanism to overcome the inhibitory effects of PARP1, promoting lytic replication.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Humans , Complement Factor D/genetics , Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Promoter Regions, Genetic , Virus Replication/genetics , Poly(ADP-ribose) Polymerases/metabolism
6.
PLoS Pathog ; 17(1): e1009261, 2021 01.
Article in English | MEDLINE | ID: mdl-33508027

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV), which belongs to the gammaherpesvirus subfamily, is associated with the pathogenesis of various tumors. Nuclear enzyme poly(ADP-ribose) polymerase 1 (PARP1) catalyzes the polymerization of ADP-ribose units on target proteins. In KSHV-infected cells, PARP1 inhibits replication and transcription activator (RTA), a molecular switch that initiates lytic replication, through direct interaction. Thus, for efficient replication, KSHV has to overcome the molecular barrier in the form of PARP1. Previously, we have demonstrated that KSHV downregulates the expression of PARP1 through PF-8, a viral processivity factor. PF-8 induces ubiquitin-proteasome system-mediated degradation of PARP1 via direct physical association and enhances RTA transactivation activity. Here, we showed that dimerization domains of PF-8 are crucial not only for PARP1 interaction and degradation but also for enhancement of the RTA transactivation activity. PF-8 recruited CHFR for the PARP1 degradation. A knockdown of CHFR attenuated the PF-8-induced PARP1 degradation and enhancement of the RTA transactivation activity, leading to reduced KSHV lytic replication. These findings reveal a mechanism by which KSHV PF-8 recruits a cellular E3 ligase to curtail the inhibitory effect of PARP1 on KSHV lytic replication.


Subject(s)
Cell Cycle Proteins/metabolism , Herpesvirus 8, Human/genetics , Immediate-Early Proteins/metabolism , Neoplasm Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Cell Cycle Proteins/genetics , Dimerization , Down-Regulation , Herpesvirus 8, Human/physiology , Humans , Immediate-Early Proteins/genetics , Neoplasm Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Protein Domains , Proteolysis , Trans-Activators/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Viral Proteins/genetics , Virus Replication
7.
Front Microbiol ; 12: 811671, 2021.
Article in English | MEDLINE | ID: mdl-35095818

ABSTRACT

The gammaherpesviruses, include the Epstein-Barr virus, Kaposi's sarcoma-associated herpesvirus, and murine gammaherpesvirus 68. They establish latent infection in the B lymphocytes and are associated with various lymphoproliferative diseases and tumors. The poly (ADP-ribose) polymerase-1 (PARP1), also called ADP-ribosyltransferase diphtheria-toxin-like 1 (ARTD1) is a nuclear enzyme that catalyzes the transfer of the ADP-ribose moiety to its target proteins and participates in important cellular activities, such as the DNA-damage response, cell death, transcription, chromatin remodeling, and inflammation. In gammaherpesvirus infection, PARP1 acts as a key regulator of the virus life cycle: lytic replication and latency. These viruses also develop various strategies to regulate PARP1, facilitating their replication. This review summarizes the roles of PARP1 in the viral life cycle as well as the viral modulation of host PARP1 activity and discusses the implications. Understanding the interactions between the PARP1 and oncogenic gammaherpesviruses may lead to the identification of effective therapeutic targets for the associated diseases.

8.
Anal Biochem ; 612: 113952, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32926865

ABSTRACT

During the development of a specific dipeptidyl peptidase 4 (DPP4) inhibitor to treat type 2 diabetes, a fluorogenic kinetic analysis for DPP4 enzymatic activity using Gly-Pro-Aminomethylcoumarin (AMC) as a substrate was optimized and validated for recombinant DPP4 and human plasma samples. The sensitivity, calibration curve, detection range, accuracy, precision, recovery efficiency, Km constant, short/long-term stability, and stability after freezing-thawing cycles were analyzed. DPP4 enzymatic activity (mU/min) was measured as the initial velocity (Vo) of the enzymatic reaction over time. The sensitivity of the Vo value was 14,488 mU/min for recombinant DPP4 and 17,995 mU/min for human plasma samples. The dynamic ranges of the calibration curve were linear and reliable between 1.11 × 104-1.86 × 106 mU/min of the mean Vo value and in the DPP4 concentration range of 23.4-3,000 ng/mL. The assay's accuracy and precision met acceptance criteria for all samples. Plasma DPP4 was stable under various storage temperatures, even after three freeze-thaw cycles. Our optimized, validated bioanalytic method for measuring DPP4 activity in plasma samples was successfully employed to evaluate the effect of evogliptin (DA-1229) tartrate, which irreversibly and dose-dependently inhibits DPP4 enzymatic activity, without the dilution effect of human plasma samples and irrespective of the co-treated metformin.


Subject(s)
Dipeptidyl Peptidase 4/blood , Enzyme Assays/methods , Spectrometry, Fluorescence/methods , Calibration , Coumarins/metabolism , Dipeptidyl Peptidase 4/analysis , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Humans , Kinetics , Limit of Detection , Piperazines/administration & dosage , Piperazines/metabolism , Piperazines/pharmacology , Protein Stability
9.
Biomol Ther (Seoul) ; 29(2): 154-165, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33148870

ABSTRACT

This study aimed to investigate whether the antidiabetic drugs dipeptidyl peptidase 4 (DPP4) inhibitors such as evogliptin and sitagliptin affect the membrane DPP4 (mDPP4) enzymatic activity and immune function of T helper1 (Th1) cells in terms of cytokine expression and cell profiles. The mDPP4 enzymatic activity, cytokine expression, and cell profiles, including cell counts, cell viability, DNA synthesis, and apoptosis, were measured in pokeweed mitogen (PWM)-activated CD4+CD26+ H9 Th1 cells with or without the DPP4 inhibitors, evogliptin and sitagliptin. PWM treatment alone strongly stimulated the expression of mDPP4 and cytokines such as interleukin (IL)-2, IL-10, tumor necrosis factor-alpha, interferon-gamma, IL-13, and granulocyte-macrophage colony stimulating factor in the CD4+CD26+ H9 Th1 cells. Evogliptin or sitagliptin treatment potently inhibited mDPP4 activity in a dose-dependent manner but did not affect either the cytokine profile or cell viability in PWM-activated CD4+CD26+ H9 Th1 cells. These results suggest that, following immune stimulation, Th1 cell signaling pathways for cytokine expression function normally after treatment with evogliptin or sitagliptin, which efficiently inhibit mDPP4 enzymatic activity in Th1 cells.

10.
Antibiotics (Basel) ; 9(10)2020 Oct 14.
Article in English | MEDLINE | ID: mdl-33066532

ABSTRACT

Noroviruses are the leading cause of acute gastroenteritis and food poisoning worldwide. In this study, we investigated the anti-noroviral activity of Lindera obtusiloba leaf extract (LOLE) using murine norovirus (MNV-1), a surrogate of human norovirus. Preincubation of MNV-1 with LOLE at 4, 8, or 12 mg/mL for 1 h at 25 °C significantly reduced viral infectivity, by 51.8%, 64.1%, and 71.2%, respectively. Among LOLE single compounds, ß-pinene (49.7%), α-phellandrene (26.2%), and (+)-limonene (17.0%) demonstrated significant inhibitory effects on viral infectivity after pretreatment with MNV-1, suggesting that the anti-noroviral effects of LOLE may be due to the synergetic activity of several compounds, with ß-pinene as a key molecule. The inhibitory effect of LOLE was tested on the edible surfaces of lettuce, cabbage, and oysters, as well as on stainless steel. After one hour of incubation at 25°C, LOLE (12 mg/mL) pretreatment significantly reduced MNV-1 plaque formation on lettuce (76.4%), cabbage (60.0%), oyster (38.2%), and stainless-steel (62.8%). These results suggest that LOLE effectively inhibits norovirus on food and metal surfaces. In summary, LOLE, including ß-pinene, may inactivate norovirus and could be used as a natural agent promoting food safety and hygiene.

11.
J Microbiol ; 58(1): 46-53, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31768937

ABSTRACT

The Chikungunya virus (CHIKV) belongs to the Alphavirus genus of Togaviridae family and contains a positive-sense single stranded RNA genome. Infection by this virus mainly causes sudden high fever, rashes, headache, and severe joint pain that can last for several months or years. CHIKV, a mosquito-borne arbovirus, is considered a re-emerging pathogen that has become one of the most pressing global health concerns due to a rapid increase in epidemics. Because handling of CHIKV is restricted to Biosafety Level 3 (BSL-3) facilities, the evaluation of prophylactic vaccines or antivirals has been substantially hampered. In this study, we first iden-tified the whole structural polyprotein sequence of a CHIKV strain isolated in South Korea (KNIH/2009/77). Phylogenetic analysis showed that this sequence clustered within the East/ Central/South African CHIKV genotype. Using this sequence information, we constructed a CHIKV-pseudotyped lenti-virus expressing the structural polyprotein of the Korean CHIKV isolate (CHIKVpseudo) and dual reporter genes of green fluorescence protein and luciferase. We then developed a pseudovirus-based neutralization assay (PBNA) using CHIKVpseudo. Results from this assay compared to those from the conventional plaque reduction neutralization test showed that our PBNA was a reliable and rapid method to evaluate the efficacy of neutralizing antibodies. More importantly, the neutralizing activities of human sera from CHIKV-infected individuals were quantitated by PBNA using CHIKVpseudo. Taken together, these results suggest that our PBNA for CHIKV may serve as a useful and safe method for testing the neutralizing activity of antibodies against CHIKV in BSL-2 facilities.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chikungunya Fever , Chikungunya virus/immunology , Neutralization Tests/methods , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/isolation & purification , Humans , Republic of Korea
12.
Pathogens ; 8(4)2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31658769

ABSTRACT

Licensed live attenuated vaccines have been developed to prevent varicella zoster virus (VZV) infection, which causes chickenpox and shingles. The genomic sequences of both clinical- and vaccine-derived VZV strains have been analyzed previously. To further characterize the molecular signatures and complexity of wildtype (clinical) versus attenuated (vaccine-derived) VZV-mediated host cellular responses, we performed high-throughput next generation sequencing to quantify and compare the expression patterns of mRNAs and microRNAs (miRNAs) in primary human dermal fibroblasts (HDFs) infected with wildtype (YC01 low passage) and attenuated (YC01 high passage, SuduVax, and VarilRix) VZV strains. 3D-multidimensional scaling of the differentially expressed genes demonstrated the distinct grouping of wildtype and attenuated strains. In particular, we observed that HDFs infected with attenuated strains had more differentially expressed genes (DEGs) involved in the retinoic-acid inducible gene-I-like receptor and interferon-mediated signaling pathways compared with wildtype strains. Additionally, miRNA expression patterns were profiled following the infection of HDFs with VZV. Small RNA sequencing identified that several miRNAs were upregulated, including miR-146a-5p, which has been associated with other herpesvirus infections, whereas let-7a-3p was downregulated in both wildtype and attenuated VZV-infected cells. This study identified genes and miRNAs that may be essential in VZV pathogenesis.

13.
Cell Host Microbe ; 25(4): 602-616.e7, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30902577

ABSTRACT

Establishing the balance between positive and negative innate immune mechanisms is crucial for maintaining homeostasis. Here we uncover the regulatory crosstalk between two previously unlinked innate immune receptor families: RIG-I, an anti-viral cytosolic receptor activated type I interferon production, and NLR (nucleotide-binding domain, leucine repeat domain-containing protein). We show that NLRP12 dampens RIG-I-mediated immune signaling against RNA viruses by controlling RIG-I's association with its adaptor MAVS. The nucleotide-binding domain of NLRP12 interacts with the ubiquitin ligase TRIM25 to prevent TRIM25-mediated, Lys63-linked ubiquitination and activation of RIG-I. NLRP12 also enhances RNF125-mediated, Lys48-linked degradative ubiquitination of RIG-I. Vesicular stomatitis virus (VSV) infection downregulates NLRP12 expression to allow RIG-I activation. Myeloid-cell-specific Nlrp12-deficient mice display a heightened interferon and TNF response and are more resistant to VSV infection. These results indicate that NLRP12 functions as a checkpoint for anti-viral RIG-I activation.


Subject(s)
DEAD Box Protein 58/immunology , DNA-Binding Proteins/immunology , Intracellular Signaling Peptides and Proteins/immunology , RNA Virus Infections/immunology , RNA Viruses/physiology , Transcription Factors/immunology , Animals , DEAD Box Protein 58/genetics , DNA-Binding Proteins/genetics , Female , Humans , Interferons/genetics , Interferons/immunology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Protein Binding , RNA Virus Infections/genetics , RNA Virus Infections/virology , RNA Viruses/genetics , Transcription Factors/genetics , Ubiquitination
14.
Biochem Biophys Res Commun ; 509(2): 414-420, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30594400

ABSTRACT

BST2 is an antiviral factor that inhibits the release of enveloped virus at the plasma membrane via an unusual topology in which its N-terminal is in the cytosol while its C-terminal is anchored by glycophosphatidylinositol (GPI). BST2-deficient cells showed substantially higher release of virions than wild type cells. Influenza-infected BST2-deficient cells showed greatly reduced cytopathic effect (CPE) than wild type cells despite their generally robust virus production. This finding prompted us to determine whether BST2 was involved in the apoptotic process of virus-infected host cells. Our results revealed that BST2 might be involved in IRE1α-mediated ER stress pathway by increasing spliced form XBP-1. Consequently, levels of cytochrome C, caspase-3, caspase-9, and PARP as representative molecules of apoptosis were significantly increased in wild type cells than those in BST2-deficient cells. These results suggest that BST2 might participate in innate host defense by augmenting ER-stress-induced apoptotic signaling to inhibit the replication and spread of virus.


Subject(s)
Antigens, CD/genetics , Endoribonucleases/genetics , Host-Pathogen Interactions/genetics , Influenza A Virus, H1N1 Subtype/genetics , Protein Serine-Threonine Kinases/genetics , X-Box Binding Protein 1/genetics , Animals , Antigens, CD/immunology , Apoptosis/genetics , Apoptosis/immunology , Caspase 3/genetics , Caspase 3/immunology , Caspase 9/genetics , Caspase 9/immunology , Chlorocebus aethiops , Cytochromes c/genetics , Cytochromes c/immunology , Dogs , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Madin Darby Canine Kidney Cells , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/immunology , Protein Serine-Threonine Kinases/immunology , Signal Transduction , Vero Cells , Virus Replication , X-Box Binding Protein 1/immunology
15.
IUCrJ ; 5(Pt 6): 866-879, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30443370

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP-1), an enzyme that modifies nuclear proteins by poly(ADP-ribosyl)ation, regulates various cellular activities and restricts the lytic replication of oncogenic gammaherpesviruses by inhibiting the function of replication and transcription activator (RTA), a key switch molecule of the viral life cycle. A viral PARP-1-interacting protein (vPIP) encoded by murine gammaherpesvirus 68 (MHV-68) orf49 facilitates lytic replication by disrupting interactions between PARP-1 and RTA. Here, the structure of MHV-68 vPIP was determined at 2.2 Šresolution. The structure consists of 12 α-helices with characteristic N-terminal ß-strands (Nß) and forms a V-shaped-twist dimer in the asymmetric unit. Structure-based mutagenesis revealed that Nß and the α1 helix (residues 2-26) are essential for the nuclear localization and function of vPIP; three residues were then identified (Phe5, Ser12 and Thr16) that were critical for the function of vPIP and its interaction with PARP-1. A recombinant MHV-68 harboring mutations of these three residues showed severely attenuated viral replication both in vitro and in vivo. Moreover, ORF49 of Kaposi's sarcoma-associated herpesvirus also directly interacted with PARP-1, indicating a conserved mechanism of action of vPIPs. The results elucidate the novel molecular mechanisms by which oncogenic gammaherpesviruses overcome repression by PARP-1 using vPIPs.

16.
J Dairy Sci ; 101(12): 10675-10684, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30316596

ABSTRACT

Influenza A virus (IAV) infection is a global public health concern. It causes respiratory diseases ranging from mild illness to fatal disease. Natural killer (NK) cells are an innate immune component that kill infected cells and secrete cytokines to modulate the adaptive immune system; they constitute the first-line defense and play important roles in controlling IAV infection. This study evaluated the effect of daily administration of heat-treated Lactobacillus plantarum nF1-fortified yogurt on immunity and protection against IAV infection. Mice administered with heat-treated L. plantarum nF1-fortified yogurt showed elevated NK cell-related cytokine expression levels. Daily administration of the L. plantarum nF1-fortified yogurt before IAV infection also enhanced splenic NK activity, lung inflammatory cytokine responses, and survival rate. Thus, daily administration of nF1-fortified yogurt enhances host immunity and helps prevent IAV infection.


Subject(s)
Influenza, Human/diet therapy , Killer Cells, Natural/immunology , Lactobacillus plantarum/chemistry , Yogurt/microbiology , Animals , Cytokines/immunology , Hot Temperature , Humans , Influenza, Human/genetics , Influenza, Human/immunology , Lactobacillus plantarum/metabolism , Male , Mice , Mice, Inbred BALB C , Spleen/immunology , Yogurt/analysis
18.
Nat Commun ; 9(1): 3284, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30115930

ABSTRACT

Cytokines are involved in early host defense against pathogen infections. In particular, tumor necrosis factor (TNF) and interferon-gamma (IFN-γ) have critical functions in non-cytopathic elimination of hepatitis B virus (HBV) in hepatocytes. However, the molecular mechanisms and mediator molecules are largely unknown. Here we show that interleukin-32 (IL-32) is induced by TNF and IFN-γ in hepatocytes, and inhibits the replication of HBV by acting intracellularly to suppress HBV transcription and replication. The gamma isoform of IL-32 (IL-32γ) inhibits viral enhancer activities by downregulating liver-enriched transcription factors. Our data are validated in both an in vivo HBV mouse model and primary human hepatocytes. This study thus suggests that IL-32γ functions as intracellular effector in hepatocytes for suppressing HBV replication to implicate a possible mechanism of non-cytopathic viral clearance.


Subject(s)
Antiviral Agents/metabolism , Cytokines/metabolism , Hepatitis B virus/physiology , Interleukins/metabolism , Intracellular Space/metabolism , Animals , Base Sequence , Cell Line, Tumor , Disease Models, Animal , Down-Regulation , Enhancer Elements, Genetic/genetics , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/pathology , Hepatocyte Nuclear Factors/metabolism , Humans , MAP Kinase Signaling System , Male , Mice , Models, Biological , Protein Binding , Transcription, Genetic , Virus Replication
19.
Nucleic Acids Res ; 46(17): 9011-9026, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30137501

ABSTRACT

DExD/H-box helicase 9 (DHX9), or RNA helicase A (RHA), is an abundant multifunctional nuclear protein. Although it was previously reported to act as a cytosolic DNA sensor in plasmacytoid dendritic cells (pDCs), the role and molecular mechanisms of action of DHX9 in cells that are not pDCs during DNA virus infection are not clear. Here, a macrophage-specific knockout and a fibroblast-specific knockdown of DHX9 impaired antiviral innate immunity against DNA viruses, leading to increased virus replication. DHX9 enhanced NF-κB-mediated transactivation in the nucleus, which required its ATPase-dependent helicase (ATPase/helicase) domain, but not the cytosolic DNA-sensing domain. In addition, DNA virus infection did not induce cytoplasmic translocation of nuclear DHX9 in macrophages and fibroblasts. Nuclear DHX9 was associated with a multiprotein complex including both NF-κB p65 and RNA polymerase II (RNAPII) in chromatin containing NF-κB-binding sites. DHX9 was essential for the recruitment of RNAPII rather than NF-κB p65, to the corresponding promoters; this function also required its ATPase/helicase activity. Taken together, our results show a critical role of nuclear DHX9 (as a transcription coactivator) in the stimulation of NF-κB-mediated innate immunity against DNA virus infection, independently of DHX9's DNA-sensing function.


Subject(s)
DEAD-box RNA Helicases/genetics , DNA, Viral/genetics , Host-Pathogen Interactions/genetics , Immunity, Innate , NF-kappa B/genetics , RNA Polymerase II/genetics , Animals , Chlorocebus aethiops , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/immunology , DNA, Viral/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Gammaherpesvirinae/genetics , Gammaherpesvirinae/growth & development , Gammaherpesvirinae/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/immunology , Host-Pathogen Interactions/immunology , Humans , Macrophages/immunology , Macrophages/virology , Male , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/immunology , Mouse Embryonic Stem Cells/virology , NF-kappa B/immunology , NIH 3T3 Cells , Primary Cell Culture , RNA Polymerase II/immunology , Signal Transduction , Vero Cells , Virus Replication
20.
PLoS One ; 13(7): e0200624, 2018.
Article in English | MEDLINE | ID: mdl-29990347

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0126456.].

SELECTION OF CITATIONS
SEARCH DETAIL
...