Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Biochem Biophys Res Commun ; 718: 149931, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38723415

ABSTRACT

Oncolytic viruses (OVs) have shown potential in converting a "cold" tumor into a "hot" one and exhibit effectiveness in various cancer types. However, only a subset of patients respond to oncolytic virotherapy. It is important to understand the resistance mechanisms to OV treatment in pancreatic ductal adenocarcinoma (PDAC) to engineer oncolytic viruses. In this study, we used transcriptome RNA sequencing (RNA-seq) to identify Visfatin, which was highly expressed in the responsive tumors following OV treatment. To explore the antitumor efficacy, we modified OV-mVisfatin, which effectively inhibited tumor growth. For the first time, we revealed that Visfatin promoted the antitumor efficacy of OV by remodeling the tumor microenvironment, which involved enhancing CD8+ T cell and DC cell infiltration and activation, repolarizing macrophages towards the M1-like phenotype, and decreasing Treg cells using single-cell RNA sequencing (scRNA-seq) and flow cytometry. Furthermore, PD-1 blockade significantly enhanced OV-mVisfatin antitumor efficacy, offering a promising new therapeutic strategy for PDAC.


Subject(s)
Herpesvirus 1, Human , Nicotinamide Phosphoribosyltransferase , Oncolytic Virotherapy , Oncolytic Viruses , Pancreatic Neoplasms , Tumor Microenvironment , Animals , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Mice , Oncolytic Virotherapy/methods , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Herpesvirus 1, Human/genetics , Cell Line, Tumor , Oncolytic Viruses/genetics , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Mice, Inbred C57BL , Humans , CD8-Positive T-Lymphocytes/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Female
2.
Biochem Biophys Res Commun ; 698: 149546, 2024 02 26.
Article in English | MEDLINE | ID: mdl-38266314

ABSTRACT

The low clinical response rate of checkpoint blockades, such as PD-1 and CTLA-4, highlighted the requirements of agonistic antibodies to boost optimal T cell responses. OX40, a co-stimulatory receptor on the T cells, plays a crucial role in promoting T cell survival and differentiation. However, the clinical efficacy of anti-OX40 agonistic antibodies was unimpressive. To explore the mechanism underlying the action of anti-OX40 agonists to improve the anti-tumor efficacy, we analyzed the dynamic changes of tumor-infiltrating immune cells at different days post-treatments using single-cell RNA-sequencing (scRNA-seq). In this study, we found that tumor-infiltrating regulatory T (Treg) cells were reduced after two rounds of anti-OX40 treatment, but the increase of infiltration and activation of CD8+ effector T cells, as well as M1 polarization in the tumor were only observed after three rounds of treatments. Moreover, our group first analyzed the antitumor effect of anti-OX40 treatments on regulating the macrophages and discovered the dynamic changes of vascular endothelial growth factor (VEGF) and CD40 signaling pathways on macrophages, indicating their possibility to being potential combination targets to improve the anti-OX40 agonists efficacy. The combination of VEGFR inhibitors or anti-CD40 agonist antibody with anti-OX40 agonists exhibited more remarkable inhibition of tumor growth. Therefore, the mechanism-driven combination of anti-OX40 agonists with VEGFR inhibitors or anti-CD40 agonists represented promising strategies.


Subject(s)
T-Lymphocytes, Regulatory , Vascular Endothelial Growth Factor A , Antibodies , Immunotherapy , Macrophages
3.
Mol Ther ; 29(2): 744-761, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33130310

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the major type of pancreatic malignancy with very poor prognosis. Despite the promising results of immune checkpoint inhibitors (ICIs) in some solid tumors, immunotherapy is less effective for PDAC due to its immunosuppressive tumor microenvironment (TME). In this report, we established an immunocompetent syngeneic PDAC model and investigated the effect of oncolytic herpes simplex virus-1 (oHSV) on the composition of TME immune cells. The oHSV treatment significantly reduced tumor burden and prolonged the survival of tumor-bearing mice. Further, by single cell RNA sequencing (scRNA-seq) and multicolor fluorescence-activated cell sorting (FACS) analysis, we demonstrated that oHSV administration downregulated tumor-associated macrophages (TAMs), especially the anti-inflammatory macrophages, and increased the percentage of tumor-infiltrating lymphocytes, including activated cytotoxic CD8+ T cells and T helper (Th)1 cells. Besides, the combination of oHSV and immune checkpoint modulators extended the lifespan of the tumor-bearing mice. Overall, our data suggested that oHSV reshapes the TME of PDAC by boosting the immune activity and leads to improved responsiveness of PDAC to immunotherapy.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Host Microbial Interactions/immunology , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Simplexvirus/genetics , Tumor Microenvironment/immunology , Animals , Biomarkers , Cytokines/metabolism , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Oncolytic Virotherapy/methods
4.
Biochem Biophys Res Commun ; 511(4): 787-793, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30833082

ABSTRACT

Vav1 is a guanine nucleotide exchange factor (GEF) predominantly expressed in hematopoietic cells, and functions in the development and antigen-stimulated response of lymphocytes. Burkitt's lymphoma (BL) is characterized as transformed B cell lymphoma, and is highly associated with Epstein-Barr virus (EBV). EBV nuclear antigen 1 (EBNA1) is the only viral protein expressed across all three types of latency and essential for the persistence of EBV genome. It is not clear yet how EBNA1 contributes to the growth advantage of latently infected cells such as in EBV+ lymphoma B cells. Here, we reported that Vav1 interacts with EBNA1 via its C-terminal SH3 domain. This interaction suppresses the expression of a pro-apoptotic Bcl-2 family member, Bim, resulting in the resistance of the BL cells to apoptotic inductions. Our data uncovered Vav1 as a novel target for EBNA1, and suggested a pro-survival role of Vav1 in the pathogenesis of EBV associated BLs.


Subject(s)
Bcl-2-Like Protein 11/genetics , Burkitt Lymphoma/metabolism , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Burkitt Lymphoma/genetics , Burkitt Lymphoma/virology , Cell Line, Tumor , Cell Survival , Down-Regulation , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Humans , Protein Interaction Maps
5.
Biochem Biophys Res Commun ; 509(4): 954-959, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30648553

ABSTRACT

Venom peptides are an excellent source of pharmacologically active molecules for ion channels that have been considered as promising drug targets. However, mining venoms that interact with ion channel remains challenging. Previously an autocrine based high throughput selection system was developed to screen venom peptide library but the method includes repetitious selection rounds that may cause loss of valuable hits. To simplify the selection process, next generation sequencing was employed to directly identify the positive hits after a single round of selection. The advantage of the improved system was demonstrated by the discovery of 3 novel Kv1.3 targeting venom peptides among which Kappa-thalatoxin-Tas2a is a potent Kv1.3 antagonist. Therefore, this simplified method is efficient to identify novel venom peptides that target ion channels.


Subject(s)
Drug Discovery , Kv1.3 Potassium Channel/antagonists & inhibitors , Peptides/analysis , Scorpion Venoms/chemistry , Animals , Autocrine Communication , High-Throughput Nucleotide Sequencing , Humans , Scorpions/pathogenicity
6.
J Virol ; 92(20)2018 10 15.
Article in English | MEDLINE | ID: mdl-30045990

ABSTRACT

The γ134.5 gene of herpes simplex virus 1 (HSV-1) encodes a virulence factor that promotes viral pathogenesis. Although it perturbs TANK-binding kinase 1 (TBK1) in the complex network of innate immune pathways, the underlying mechanism is obscure. Here we report that HSV-1 γ134.5 targets stimulator of interferon genes (STING) in the intracellular DNA recognition pathway that regulates TBK1 activation. In virus-infected cells the γ134.5 protein associates with and inactivates STING, which leads to downregulation of interferon regulatory factor 3 (IRF3) and IFN responses. Importantly, HSV-1 γ134.5 disrupts translocation of STING from the endoplasmic reticulum to Golgi apparatus, a process necessary to prime cellular immunity. Deletion of γ134.5 or its amino-terminal domain from HSV-1 abolishes the observed inhibitory activities. Consistently, an HSV mutant that lacks functional γ134.5 replicated less efficiently in STING+/+ than in STING-/- mouse embryonic fibroblasts. Moreover, reconstituted expression of human STING in the STING-/- cells activated IRF3 and reduced viral growth. These results suggest that control of the DNA sensing pathway by γ134.5 is advantageous to HSV infection.IMPORTANCE Viral inhibition of innate immunity contributes to herpes simplex virus pathogenesis. Although this complex process involves multiple factors, the underlying events remain unclear. We demonstrate that an HSV virulence factor γ134.5 precludes the activation of STING, a central adaptor in the intracellular DNA sensing pathway. Upon HSV infection, this viral protein engages with and inactivates STING. Consequently, it compromises host immunity and facilitates HSV replication. These observations uncover an HSV mechanism that is likely to mediate viral virulence.


Subject(s)
Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Immune Evasion , Membrane Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Virulence Factors/metabolism , Virus Replication , Animals , Cell Line , Chlorocebus aethiops , Down-Regulation , Gene Deletion , Genetic Complementation Test , Herpesvirus 1, Human/immunology , Humans , Interferon Regulatory Factor-3/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Viral Proteins/genetics , Virulence Factors/genetics
7.
J Lipid Res ; 59(12): 2287-2296, 2018 12.
Article in English | MEDLINE | ID: mdl-30309895

ABSTRACT

Production of 25-hydroxycholesterol (25HC), a potent inhibitor of viral infection, is catalyzed by cholesterol 25-hydroxylase (CH25H). We previously reported that 25HC induced CH25H expression in a liver X receptor (LXR)-dependent manner, implying that LXR can play an important role in antiviral infection. In this study, we determined that activation of LXR by 25HC or synthetic ligands [T0901317 (T317) or GW3965] inhibited infection of herpes simplex virus type 1 (HSV-1) or MLV-(VSV)-GFP in HepG2 cells or RAW 264.7 macrophages. Genetic deletion of LXRα, LXRß, or CH25H expression in HepG2 cells by CRISPR/Cas9 method increased cell susceptibility to HSV-1 infection and attenuated the inhibition of LXR on viral infection. Lack of interferon (IFN)-γ expression also increased cell susceptibility to viral infection. However, it attenuated, but did not block, the inhibition of LXR on HSV-1 infection. In addition, expression of CH25H, but not IFN-γ, was inversely correlated to cell susceptibility to viral infection and the antiviral actions of LXR. Metabolism of 25HC into 25HC-3-sulfate (25HC3S) by cholesterol sulfotransferase-2B1b moderately reduced the antiviral actions of 25HC because 25HC3S is a weaker inhibitor of HSV-1 infection than 25HC. Furthermore, administration of T317 to BALB/c mice reduced HSV-1 growth in mouse tissues. Taken together, we demonstrate an antiviral system of 25HC with involvement of LXR activation, interaction between CH25H and IFN-γ, and 25HC metabolism.


Subject(s)
Hydroxycholesterols/metabolism , Liver X Receptors/metabolism , Animals , Blotting, Western , CRISPR-Cas Systems/genetics , Hep G2 Cells , Herpesvirus 1, Human/metabolism , Humans , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Sulfotransferases/metabolism
8.
J Virol ; 90(22): 10414-10422, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27630226

ABSTRACT

Herpes simplex virus 1 (HSV-1) remodels nuclear membranes during virus egress. Although the UL31 and UL34 proteins control nucleocapsid transit in infected cells, the molecular interactions required for their function are unclear. Here we report that the γ134.5 gene product of HSV-1 facilitates nucleocapsid release to the cytoplasm through bridging the UL31/UL34 complex, cellular p32, and protein kinase C. Unlike wild-type virus, an HSV mutant devoid of γ134.5 or its amino terminus is crippled for viral growth and release. This is attributable to a defect in virus nuclear egress. In infected cells, wild-type virus recruits protein kinase C to the nuclear membrane and triggers its activation, whereas the γ134.5 mutants fail to exert such an effect. Accordingly, the γ134.5 mutants are unable to induce phosphorylation and reorganization of lamin A/C. When expressed in host cells γ134.5 targets p32 and protein kinase C. Meanwhile, it communicates with the UL31/UL34 complex through UL31. Deletion of the amino terminus from γ134.5 disrupts its activity. These results suggest that disintegration of the nuclear lamina mediated by γ134.5 promotes HSV replication. IMPORTANCE: HSV nuclear egress is a key step that determines the outcome of viral infection. While the nuclear egress complex mediates capsid transit across the nuclear membrane, the regulatory components are not clearly defined in virus-infected cells. We report that the γ134.5 gene product, a virulence factor of HSV-1, facilitates nuclear egress cooperatively with cellular p32, protein kinase C, and the nuclear egress complex. This work highlights a viral mechanism that may contribute to the pathogenesis of HSV infection.


Subject(s)
Herpesvirus 1, Human/metabolism , Lamin Type A/metabolism , Phosphorylation/physiology , Viral Proteins/metabolism , Virus Release/physiology , Animals , Capsid/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/virology , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoplasm/virology , HeLa Cells , Humans , Nuclear Envelope/metabolism , Nuclear Envelope/virology , Nuclear Lamina/metabolism , Nuclear Lamina/virology , Nuclear Proteins/metabolism , Nucleocapsid/metabolism , Protein Kinase C/metabolism , Vero Cells , Virus Assembly/physiology
9.
J Biol Chem ; 290(25): 15670-15678, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25907557

ABSTRACT

Herpes simplex virus 1 (HSV-1) is the most prevalent human virus and causes global morbidity because the virus is able to infect multiple cell types. Remarkably, HSV infection switches between lytic and latent cycles, where T cells play a critical role. However, the precise way of virus-host interactions is incompletely understood. Here we report that HSV-1 productively infected Jurkat T-cells and inhibited antigen-induced T cell receptor activation. We discovered that HSV-1-encoded Us3 protein interrupted TCR signaling and interleukin-2 production by inactivation of the linker for activation of T cells. This study unveils a mechanism by which HSV-1 intrudes into early events of TCR-mediated cell signaling and may provide novel insights into HSV infection, during which the virus escapes from host immune surveillance.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Membrane Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 6/immunology , Viral Proteins/immunology , Adaptor Proteins, Signal Transducing/genetics , Herpes Simplex/genetics , Herpes Simplex/pathology , Herpesvirus 1, Human/genetics , Humans , Immune Evasion/genetics , Interleukin-2/genetics , Interleukin-2/immunology , Jurkat Cells , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell , Signal Transduction/genetics , T-Lymphocytes/pathology , T-Lymphocytes/virology , TNF Receptor-Associated Factor 6/genetics , Viral Proteins/genetics
10.
J Biol Chem ; 289(52): 35795-805, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25355318

ABSTRACT

As a large double-stranded DNA virus, herpes simplex virus type 1 (HSV-1) assembles capsids in the nucleus where the viral particles exit by budding through the inner nuclear membrane. Although a number of viral and host proteins are involved, the machinery of viral egress is not well understood. In a search for host interacting proteins of ICP34.5, which is a virulence factor of HSV-1, we identified a cellular protein, p32 (gC1qR/HABP1), by mass spectrophotometer analysis. When expressed, ICP34.5 associated with p32 in mammalian cells. Upon HSV-1 infection, p32 was recruited to the inner nuclear membrane by ICP34.5, which paralleled the phosphorylation and rearrangement of nuclear lamina. Knockdown of p32 in HSV-1-infected cells significantly reduced the production of cell-free viruses, suggesting that p32 is a mediator of HSV-1 nuclear egress. These observations suggest that the interaction between HSV-1 ICP34.5 and p32 leads to the disintegration of nuclear lamina and facilitates the nuclear egress of HSV-1 particles.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/virology , Herpesvirus 1, Human/physiology , Mitochondrial Proteins/metabolism , Viral Proteins/metabolism , Animals , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Protein Interaction Mapping , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Vero Cells , Virus Release , Lamin B Receptor
11.
Biochem Biophys Res Commun ; 456(1): 434-9, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25482447

ABSTRACT

Parthenolide (PTL) is a sesquiterpene lactone isolated from feverfew and exhibits potent antitumor activity against various cancers. Many studies indicate that PTL treatment leads to apoptosis, however, the mechanism has not been defined. Here, we observed that cells underwent autophagy shortly after PTL treatment. Inhibition of autophagy by knocking out autophagy associated gene atg5 blocked PTL-induced apoptosis. Surprisingly, PTL decreased the level of translation initiation factor eIF4E binding protein 1 (4E-BP1) in correlation with autophagy. Ectopic expression or shRNA knockdown of 4E-BP1 further verified the effect of 4E-BP1 on PTL-induced autophagy. Meanwhile, PTL elevated the cellular reactive oxygen species (ROS) which located upstream of the depletion of 4E-BP1, and contributed to the consequent autophagy. This study revealed 4E-BP1 as a trigger for PTL-induced autophagy and may lead to therapeutic strategy to enhance the efficacy of anticancer drugs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Autophagy/drug effects , Carrier Proteins/metabolism , Phosphoproteins/metabolism , Sesquiterpenes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle Proteins , Eukaryotic Initiation Factors , Fibroblasts/metabolism , HEK293 Cells , HL-60 Cells , HeLa Cells , Humans , Mice , Phagosomes/metabolism , Phosphorylation/drug effects , Plasmids , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
13.
J Biol Chem ; 288(6): 3777-85, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23271736

ABSTRACT

Vav1 is a guanine nucleotide exchange factor (GEF) specifically expressed in hematopoietic cells. It consists of multiple structural domains and plays important roles in T cell activation. The other highly conserved isoforms of Vav family, Vav2 and Vav3, are ubiquitously expressed in human tissues including lymphocytes. All three Vav proteins activate Rho family small GTPases, which are involved in a variety of biological processes during T cell activation. Intensive studies have demonstrated that Vav1 is indispensable for T cell receptor (TCR)-mediated signal transduction, whereas Vav2 and Vav3 function as GEFs that overlap with Vav1 on TCR-induced cytoskeleton reorganization. T cells lacking Vav1 exhibited severe defect in TCR-mediated calcium elevation, indicating that the co-existing Vav2 and Vav3 did not compensate Vav1 in calcium signaling. What is the functional particularity of Vav1 in lymphocytes? In this study, we identified the N-terminal 20 amino acids of Vav1 in the calponin homology (CH) domain to be essential for its interaction with calmodulin (CaM) that leads to TCR-induced calcium mobilization. Substitution of the 1-20 amino acids of Vav1 with those of Vav2 or Vav3 abolished the association with CaM, and the N-terminal mutations of Vav1 failed to potentiate normal TCR-induced calcium mobilization, that in turn, suspended nuclear factor of activated T cells (NFAT) activation and IL-2 production. This study highlights the importance of the N-terminal 20 aa of Vav1 for CaM binding, and provides new insights into the distinguished and irreplaceable role of Vav1 in T cell activation and signal transduction.


Subject(s)
Calcium Signaling/physiology , Lymphocyte Activation/physiology , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Calmodulin/genetics , Calmodulin/metabolism , HeLa Cells , Humans , Interleukin-2/biosynthesis , Interleukin-2/genetics , Mutation , Protein Binding/physiology , Protein Isoforms , Protein Structure, Tertiary , Proto-Oncogene Proteins c-vav/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology
14.
J Infect Dis ; 208(8): 1294-304, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23847059

ABSTRACT

BACKGROUND: γδ T cells comprise a small subset of T cells and play a protective role against cancer and viral infections; however, their precise role in patients with chronic hepatitis B remains unclear. METHODS: Flow cytometry and immunofunctional assays were performed to analyze the impact of Vδ2 γδ (Vδ2) T cells in 64 immune-activated patients, 22 immune-tolerant carriers, and 30 healthy controls. RESULTS: The frequencies of peripheral and hepatic Vδ2 T cells decreased with disease progression from immune tolerant to immune activated. In the latter group of patients, the decreases in peripheral and intrahepatic frequencies of Vδ2 T cells reversely correlated with alanine aminotransferase levels and histological activity index. These activated terminally differentiated memory phenotypic Vδ2 T cells exhibited impaired abilities in proliferation and chemotaxis, while maintained a relative intact interferon (IFN) γ production. Importantly, Vδ2 T cells, in vitro, significantly suppressed the production of cytokines associated with interleukin 17-producing CD4+ T (Th17) cells through both cell contact-dependent and IFN-γ-dependent mechanisms. CONCLUSIONS: Inflammatory microenvironment in IA patients result in decreased numbers of Vδ2 T cells, which play a novel role by regulating the pathogenic Th17 response to protect the liver in patients with chronic hepatitis B.


Subject(s)
Hepatitis B, Chronic/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Adolescent , Adult , Cell Growth Processes/immunology , Chemotaxis, Leukocyte , Cytokines/metabolism , Female , Flow Cytometry , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/pathology , Humans , Male , Middle Aged , Statistics, Nonparametric
15.
J Virol ; 86(4): 2188-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22171259

ABSTRACT

The γ(1)34.5 protein of herpes simplex viruses (HSV) is essential for viral pathogenesis, where it precludes translational arrest mediated by double-stranded-RNA-dependent protein kinase (PKR). Paradoxically, inhibition of PKR alone is not sufficient for HSV to exhibit viral virulence. Here we report that γ(1)34.5 inhibits TANK binding kinase 1 (TBK1) through its amino-terminal sequences, which facilitates viral replication and neuroinvasion. Compared to wild-type virus, the γ(1)34.5 mutant lacking the amino terminus induces stronger antiviral immunity. This parallels a defect of γ(1)34.5 for interacting with TBK1 and reducing phosphorylation of interferon (IFN) regulatory factor 3. This activity is independent of PKR. Although resistant to IFN treatment, the γ(1)34.5 amino-terminal deletion mutant replicates at an intermediate level between replication of wild-type virus and that of the γ(1)34.5 null mutant in TBK1(+/+) cells. However, such impaired viral growth is not observed in TBK1(-/-) cells, indicating that the interaction of γ(1)34.5 with TBK1 dictates HSV infection. Upon corneal infection, this mutant replicates transiently but barely invades the trigeminal ganglia or brain, which is a difference from wild-type virus and the γ(1)34.5 null mutant. Therefore, in addition to PKR, γ(1)34.5 negatively regulates TBK1, which contributes viral replication and spread in vivo.


Subject(s)
Down-Regulation , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Viral Proteins/metabolism , Virus Replication , Animals , Cell Line , Herpesvirus 1, Human/genetics , Humans , Interferon Regulatory Factor-3/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/genetics
16.
Virus Res ; 323: 198979, 2023 Jan 02.
Article in English | MEDLINE | ID: mdl-36283533

ABSTRACT

Oncolytic viruses are an emerging cancer treatment modality with promising results in clinical trials. The new generation of oncolytic viruses are genetically modified to enhance virus selectivity for tumor cells and allow local expression of therapeutic genes in tumors. The traditional technique for viral genome engineering based on homologous recombination using a bacterial artificial chromosome (BAC) system is laborious and time-consuming. With the advent of the CRISPR/Cas9 system, the efficiency of gene editing in human cells and other organisms has dramatically increased. In this report, we successfully applied the CRISPR/Cas9 technique to construct an HSV-based oncolytic virus, where the ICP34.5 coding region was replaced with the therapeutic genes murine interleukin 12 (IL12, p40-p35) and C-X-C motif chemokine ligand 11 (CXCL11), and ICP47 gene was deleted. The combination of IL12 and CXCL11 in oncolytic viruses showed considerable promise in colorectal cancer (CRC) treatment. Overall, our study describes genetic modification of the HSV-1 genome using the CRISPR/Cas9 system and provides evidence from principle studies for engineering of the HSV genome to express foreign genes.

17.
Theranostics ; 13(12): 4016-4029, 2023.
Article in English | MEDLINE | ID: mdl-37554264

ABSTRACT

Rationale: The resistance of pancreatic ductal adenocarcinoma (PDAC) to immunotherapies is caused by the immunosuppressive tumor microenvironment (TME) and dense extracellular matrix. Currently, the efficacy of an isolated strategy targeting stromal desmoplasia or immune cells has been met with limited success in the treatment of pancreatic cancer. Oncolytic virus (OV) therapy can remodel the TME and damage tumor cells either by directly killing them or by enhancing the anti-tumor immune response, which holds promise for the treatment of PDAC. This study aimed to investigate the therapeutic effect of OX40L-armed OV on PDAC and to elucidate the underlying mechanisms. Methods: Murine OX40L was inserted into herpes simplex virus-1 (HSV-1) to construct OV-mOX40L. Its expression and function were assessed using reporter cells, cytopathic effect, and immunogenic cell death assays. The efficacy of OV-mOX40L was then evaluated in a KPC syngeneic mouse model. Tumor-infiltrating immune and stromal cells were analyzed using flow cytometry and single-cell RNA sequencing to gain insight into the mechanisms of oncolytic virotherapy. Results: OV-mOX40L treatment delayed tumor growth in KPC tumor-bearing C57BL/6 mice. It also boosted the tumor-infiltrating CD4+ T cell response, mitigated cytotoxic T lymphocyte (CTL) exhaustion, and reduced the number of regulatory T cells. The treatment of OV-mOX40L reprogrammed macrophages and neutrophils to a more pro-inflammatory anti-tumor state. In addition, the number of myofibroblastic cancer-associated fibroblasts (CAF) was reduced after treatment. Based on single-cell sequencing analysis, OV-mOX40L, in combination with anti-IL6 and anti-PD-1, significantly extended the lifespan of PDAC mice. Conclusion: OV-mOX40L converted the immunosuppressive tumor immune microenvironment to a more activated state, remodeled the stromal matrix, and enhanced T cell response. OV-mOX40L significantly prolonged the survival of PDAC mice, either as a monotherapy or in combination with synergistic antibodies. Thus, this study provides a multimodal therapeutic strategy for pancreatic cancer treatment.


Subject(s)
Carcinoma, Pancreatic Ductal , Oncolytic Virotherapy , Oncolytic Viruses , Pancreatic Neoplasms , Animals , Mice , Tumor Microenvironment , Mice, Inbred C57BL , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms
18.
J Biol Chem ; 286(28): 24785-92, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21622569

ABSTRACT

The ICP34.5 protein of herpes simplex virus type 1 is a neurovirulence factor that plays critical roles in viral replication and anti-host responses. One of its functions is to recruit protein phosphatase 1 (PP1) that leads to the dephosphorylation of the α subunit of translation initiation factor eIF2 (eIF2α), which is inactivated by infection-induced phosphorylation. As PP1 is a protein phosphatase with a wide range of substrates, the question remains to be answered how ICP34.5 directs PP1 to specifically dephosphorylate eIF2α. Here we report that ICP34.5 not only binds PP1 but also associates with eIF2α by in vitro and in vivo assays. The binding site of eIF2α is identified at amino acids 233-248 of ICP34.5, which falls in the highly homologous region with human gene growth arrest and DNA damage 34. The interaction between ICP34.5 and eIF2α is independent of the phosphorylation status of eIF2α at serine 51. Deletion mutation of this region results in the failure of dephosphorylation of eIF2α by PP1 and, consequently, interrupts viral protein synthesis and replication. Our data illustrated that the binding between viral protein ICP34.5 and the host eIF2α is crucial for the specific dephosphorylation of eIF2α by PP1. We propose that herpes simplex virus protein ICP34.5 bridges PP1 and eIF2α via their binding motifs and thereby facilitates the protein synthesis and viral replication.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Herpesvirus 1, Human/physiology , Protein Biosynthesis , Protein Phosphatase 1/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Amino Acid Motifs , Animals , Binding Sites , Chlorocebus aethiops , Eukaryotic Initiation Factor-2/genetics , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Protein Phosphatase 1/genetics , Sequence Deletion , Vero Cells , Viral Proteins/genetics
19.
J Biol Chem ; 286(49): 42316-42324, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22009749

ABSTRACT

Phosphatases of the regenerating liver (PRL) play oncogenic roles in cancer development and metastasis. Although previous studies indicate that PRL-1 promotes cell growth and migration by activating both the ERK1/2 and RhoA pathways, the mechanism by which it activates these signaling events remains unclear. We have identified a PRL-1-binding peptide (Peptide 1) that shares high sequence identity with a conserved motif in the Src homology 3 (SH3) domain of p115 Rho GTPase-activating protein (GAP). p115 RhoGAP directly binds PRL-1 in vitro and in cells via its SH3 domain. Structural analyses of the PRL-1·Peptide 1 complex revealed a novel protein-protein interaction whereby a sequence motif within the PxxP ligand-binding site of the p115 RhoGAP SH3 domain occupies a folded groove within PRL-1. This prevents the canonical interaction between the SH3 domain of p115 RhoGAP and MEKK1 and results in activation of ERK1/2. Furthermore, PRL-1 binding activates RhoA signaling by inhibiting the catalytic activity of p115 RhoGAP. The results demonstrate that PRL-1 binding to p115 RhoGAP provides a coordinated mechanism underlying ERK1/2 and RhoA activation.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation, Enzymologic , Guanine Nucleotide Exchange Factors/chemistry , Immediate-Early Proteins/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Tyrosine Phosphatases/metabolism , src-Family Kinases/metabolism , Amino Acid Motifs , Animals , Fibroblasts/metabolism , HEK293 Cells , Humans , Ligands , Mice , Protein Binding , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , rho GTP-Binding Proteins/metabolism
20.
J Virol ; 85(7): 3397-407, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21248029

ABSTRACT

Dendritic cells are sentinels in innate and adaptive immunity. Upon virus infection, a complex program is in operation, which activates IκB kinase (IKK), a key regulator of inflammatory cytokines and costimulatory molecules. Here we show that the γ(1)34.5 protein, a virulence factor of herpes simplex viruses, blocks Toll-like receptor-mediated dendritic cell maturation. While the wild-type virus inhibits the induction of major histocompatibility complex (MHC) class II, CD86, interleukin-6 (IL-6), and IL-12, the γ(1)34.5-null mutant does not. Notably, γ(1)34.5 works in the absence of any other viral proteins. When expressed in mammalian cells, including dendritic cells, γ(1)34.5 associates with IKKα/ß and inhibits NF-κB activation. This is mirrored by the inhibition of IKKα/ß phosphorylation, p65/RelA phosphorylation, and nuclear translocation in response to lipopolysaccharide or poly(I:C) stimulation. Importantly, γ(1)34.5 recruits both IKKα/ß and protein phosphatase 1, forming a complex that dephosphorylates two serine residues within the catalytic domains of IκB kinase. The amino-terminal domain of γ(1)34.5 interacts with IKKα/ß, whereas the carboxyl-terminal domain binds to protein phosphatase 1. Deletions or mutations in either domain abolish the activity of γ(1)34.5. These results suggest that the control of IκB kinase dephosphorylation by γ(1)34.5 represents a critical viral mechanism to disrupt dendritic cell functions.


Subject(s)
Dendritic Cells/physiology , Herpesviridae/pathogenicity , I-kappa B Kinase/metabolism , Protein Phosphatase 1/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Cell Differentiation , Cells, Cultured , Humans , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Sequence Deletion , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL