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










Publication year range
1.
Environ Toxicol ; 39(3): 1294-1302, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37948429

ABSTRACT

Lead (Pb) is nonbiodegradable and toxic to the lungs. To investigate the potential mechanisms of Pb-induced reactive oxygen species (ROS) accumulation and cell death in the lungs, human non-small lung carcinoma H460 cells were stimulated with Pb(NO3 )2 in this study. The results showed that Pb(NO3 )2 stimulation increased cell death by inducing cell apoptosis which showed a reduced Bcl-2 expression and an enhanced caspase 3 activation. Pb(NO3 )2 also caused the production of H2 O2 in H460 cells that triggering the buildup of ROS and mitochondrial membrane potential loss. We found that Pb(NO3 )2 modulates oxidoreductive activity through reduced the glutathione-disulfide reductase and glutathione levels in Pb(NO3 )2 -exposed H460 cells. Furthermore, the superoxide dismutase (SOD) upstream molecule sirtuin 3 (SIRT3) was increased with Pb(NO3 )2 dose. Collectively, these results demonstrate that Pb(NO3 )2 promotes lung cell death through SIRT3/SOD-mediated ROS accumulation and mitochondrial dysfunction.


Subject(s)
Sirtuin 3 , Humans , Reactive Oxygen Species/metabolism , Sirtuin 3/metabolism , Lead , Mitochondria/metabolism , Superoxide Dismutase/metabolism , Apoptosis
2.
Life Sci ; 321: 121615, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37001403

ABSTRACT

AIMS: Actinomycin (Act) D, a polypeptide antibiotic, is used clinically to inhibit the growth of malignant tumors. Act D binds to DNA at the transcription initiation complex to prevent the elongation of RNA. Act D causes DNA damage, growth inhibition, and cell death. Myeloid cell leukemia (Mcl-1) is an anti-apoptotic Bcl-2 family member protein, and the present study explored the effects and molecular mechanism of Act D-induced Mcl-1 downregulation. MAIN METHODS: Human adenocarcinoma A549 cells were used to check the cytotoxic signaling pathways of Act D, particularly in apoptotic mechanism, in a cell-based study approach. Specific blockers targeting the apoptotic factors were examined for their possible roles. KEY FINDINGS: We found that Act D caused cell growth inhibition and apoptosis. Propidium iodide-based flow cytometric analysis and immunostaining confirmed cell apoptosis. Treatment with Act D caused DNA damage, followed by p53-independent cell death. Western blotting showed a significant decrease in Mcl-1 expression, mitochondrial transmembrane potential loss, and caspase-9/caspase-3 cascade activation. The proteasome inhibitor MG132 reversed Act D-induced Mcl-1 downregulation. However, pharmacological inhibition of glycogen synthase kinase-3, p53 expression, ER stress, autophagy, and vesicle acidification, which are Mcl-1-regulating signaling pathways, did not rescue these effects. Notably, Cullin-Ring E3 ligase partially mediated Mcl-1 downregulation. Administration of transforming growth factor-ß induced mesenchymal cell differentiation, but Act D still decreased Mcl-1 and caused cell apoptosis. SIGNIFICANCE: All of these data show a potential pro-apoptotic effect for Act D by facilitating Mcl-1 uncanonical downregulation.


Subject(s)
Leukemia , Lung Neoplasms , Humans , Dactinomycin/pharmacology , Dactinomycin/metabolism , Down-Regulation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Anti-Bacterial Agents/pharmacology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Lung Neoplasms/metabolism , Apoptosis , Myeloid Cells , Proto-Oncogene Proteins c-bcl-2/metabolism
4.
Int J Med Sci ; 19(10): 1502-1509, 2022.
Article in English | MEDLINE | ID: mdl-36185331

ABSTRACT

Fine particulate matter (PM2.5) is the critical cause of lung cancer and can further promote tumor cell migration and invasion. This study investigated the effects of luteolin, an antiangiogenic flavonoid agent, on blocking aqueous extract PM2.5-prompted cancer progression. We observed that luteolin reduced cell migration and the expression of pro-metastatic factors pro-matrix metalloproteinase (MMP)-2 and intercellular adhesion molecule (ICAM)-1 in PM2.5-exposed H460 lung cancer cells. Luteolin treatment also reduced the transduction of PM2.5-induced epidermal growth factor receptor (EGFR)-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) cascade signaling. Furthermore, the reduction of MMP-2 expression and ICAM-1 production by luteolin in PM2.5-stimulated H460 cells is EGFR-PI3K-AKT pathway dependent. These results suggest that luteolin exhibits antitumor progression by inhibiting EGFR-PI3K-AKT pathway.


Subject(s)
Lung Neoplasms , Matrix Metalloproteinase 2 , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Luteolin/pharmacology , Luteolin/therapeutic use , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Particulate Matter/toxicity , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
5.
JCI Insight ; 7(21)2022 11 08.
Article in English | MEDLINE | ID: mdl-36125898

ABSTRACT

Diabetes mellitus (DM) is highly comorbid with severe dengue diseases; however, the underlying mechanisms are unclear. Patients with DM have a 1.61-fold increased risk of developing dengue hemorrhagic fever. In search of host factors involved in dengue virus (DENV) infection, we used high-glucose (HG) treatment and showed that HG increased viral protein expression and virion release but had no effects on the early stages of viral infection. After HG stimulation, DENV-firefly luciferase-transfected assay and cellular replicon-based assay indicated increased viral translation, whereas using the glucose uptake inhibitor phloretin blocked this effect. HG treatment increased the translational factor poly(A)-binding protein (PABP) in a glucose transporter-associated, PI3K/AKT-regulated manner. Silencing PABP significantly decreased HG-prompted virion production. HG enhanced the formation of the PABP-eukaryotic translation initiation factor 4G complex, which is regulated by protein-disulfide isomerase. Hyperglycemia increased PABP expression, mortality rate, viral protein expression, and viral loads in streptozotocin-induced DM mice. Overall, hyperglycemic stress facilitates DENV infection by strengthening PABP-mediated viral translation.


Subject(s)
Dengue , Hyperglycemia , Animals , Mice , Protein Biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/metabolism , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Viral Proteins/metabolism , Hyperglycemia/complications
6.
Biochem Biophys Res Commun ; 617(Pt 2): 11-17, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35689837

ABSTRACT

Exposure to particulate matter 2.5 (PM2.5) has been linked to ocular surface diseases, yet knowledge of the molecular mechanism impacted on retina pathogenesis is limited. Therefore, the purpose of this study was to explore the effects and involved factors of PM2.5 exposure in human retinal pigment epithelial APRE-19 cells. Our data revealed a decreased cell viability and an increased migratory ability in APRE-19 cells after PM2.5 stimulation. The MMP-2 and MMP-9 protein levels were markedly increased while the MMPs regulators TIMP-1 and TIMP-2 were significantly reduced in PM2.5-exposed APRE-19 cells. PM2.5 also increased pro-MMP-2 expression in the cell culture supernatants. Additionally, PM2.5 promoted the EMT markers through the activation of PI3K/AKT/mTOR pathway. Moreover, the ICAM-1 production was also remarkably increased by PM2.5 but reduced by PI3K/AKT inhibitor LY294002 in APRE-19 cells. Taken together, these results suggest that PM2.5 promotes EMT in a PI3K/AKT/mTOR-dependent manner in the retinal pigment epithelium.


Subject(s)
Particulate Matter , Phosphatidylinositol 3-Kinases , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Humans , Particulate Matter/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigments/metabolism , Retinal Pigments/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
7.
Oxid Med Cell Longev ; 2022: 4978556, 2022.
Article in English | MEDLINE | ID: mdl-35308172

ABSTRACT

Brain inflammation, a pathological feature of neurodegenerative disorders, exhibits elevated microglial activity and increased levels of inflammatory factors. The present study was aimed at assessing the anti-inflammatory response of tetrahydrocurcumin (THC), the primary hydrogenated metabolite of curcumin, which was applied to treat Pseudomonas aeruginosa (P.a.) lipopolysaccharide- (LPS-) stimulated BV2 microglial cells. THC reduced P.a. LPS-induced mortality and the production of inflammatory mediators IL-6, TNF-α, MIP-2, IP-10, and nitrite. A further investigation revealed that THC decreased these inflammatory cytokines synergistically with JAK/STAT signaling inhibitors. THC also increased Nrf2/HO-1 signaling transduction which inhibits iNOS/COX-2/pNFκB cascades. Additionally, the presence of the HO-1 inhibitor Snpp increased the levels of IP-10, IL-6, and nitrite while THC treatment reduced those inflammatory factors in P.a. LPS-stimulated BV2 cells. In summary, we demonstrated that THC exhibits anti-inflammatory activities in P.a. LPS-induced inflammation in brain microglial cells by inhibiting STAT1/3-dependent NF-κB activation and inducing Nrf2-mediated HO-1 expression.


Subject(s)
Curcumin , Inflammation , Animals , Curcumin/analogs & derivatives , Curcumin/metabolism , Curcumin/pharmacology , Heme Oxygenase-1/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Janus Kinase 1 , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Pseudomonas aeruginosa
8.
J Immunol Res ; 2021: 7652569, 2021.
Article in English | MEDLINE | ID: mdl-34734091

ABSTRACT

Background. Dengue virus (DENV) infection is the most common arboviral disease that affects tropical and subtropical regions. Based on the clinical hallmarks, the different severities of patients range from mild dengue fever (MDF) to severe dengue diseases (SDDs) and include dengue hemorrhagic fever or dengue shock syndrome. These are commonly associated with cytokine release syndrome (CRS). The types and levels of cytokines/chemokines, which are suppressed or enhanced, are varied, indicating CRS's pathogenic and host defensive effects. Principal Finding. In this study, we created an integrated and precise multiplex panel of cytokine/chemokine assays based on our literature analysis to monitor dengue CRS. A 24-plex panel of cytokines/chemokines was evaluated to measure the plasma levels of targeting factors in dengue patients with an MDF and SDD diagnosis without or with comorbidities. As identified in sixteen kinds of cytokines/chemokines, ten were significantly (P < 0.05) (10/16) increased, one was significantly (P < 0.01) (1/16) decreased, and five were potentially (5/16) altered in all dengue patients (n = 30) in the acute phase of disease onset. Compared to MDF, the levels of IL-8 (CXCL-8) and IL-18 in SDD were markedly (P < 0.05) increased, accompanied by positively increased IL-6 and TNF-α and decreased IFN-γ and RANTES. With comorbidities, SDD significantly (P < 0.01) portrayed elevated IL-18 accompanied by increased IL-6 and decreased IFN-α2 and IL-12. In addition, decreased platelets were significantly (P < 0.05) associated with increased IL-18. Significance. These results demonstrate an efficient panel of dengue cytokine/chemokine assays used to explore the possible level of CRS during the acute phase of disease onset; also, we are the first to report the increase of IL-18 in severe dengue with comorbidity compared to severe dengue without comorbidity and mild dengue.


Subject(s)
Interleukin-18/blood , Severe Dengue/diagnosis , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Dengue Virus/immunology , Disease Progression , Female , Humans , Interleukin-18/immunology , Male , Middle Aged , Severe Dengue/blood , Severe Dengue/immunology , Severe Dengue/virology , Young Adult
9.
Cells ; 10(11)2021 11 15.
Article in English | MEDLINE | ID: mdl-34831405

ABSTRACT

Infection with flaviviruses causes mild to severe diseases, including viral hemorrhagic fever, vascular shock syndrome, and viral encephalitis. Several animal models explore the pathogenesis of viral encephalitis, as shown by neuron destruction due to neurotoxicity after viral infection. While neuronal cells are injuries caused by inflammatory cytokine production following microglial/macrophage activation, the blockade of inflammatory cytokines can reduce neurotoxicity to improve the survival rate. This study investigated the involvement of macrophage phenotypes in facilitating CNS inflammation and neurotoxicity during flavivirus infection, including the Japanese encephalitis virus, dengue virus (DENV), and Zika virus. Mice infected with different flaviviruses presented encephalitis-like symptoms, including limbic seizure and paralysis. Histology indicated that brain lesions were identified in the hippocampus and surrounded by mononuclear cells. In those regions, both the infiltrated macrophages and resident microglia were significantly increased. RNA-seq analysis showed the gene profile shifting toward type 1 macrophage (M1) polarization, while M1 markers validated this phenomenon. Pharmacologically blocking C-C chemokine receptor 2 and tumor necrosis factor-α partly retarded DENV-induced M1 polarization. In summary, flavivirus infection, such as JEV and DENV, promoted type 1 macrophage polarization in the brain associated with encephalitic severity.


Subject(s)
Cell Polarity , Dengue Virus/physiology , Encephalitis Virus, Japanese/physiology , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Macrophages/pathology , Severity of Illness Index , Animals , Animals, Suckling , Cell Line , Disease Models, Animal , Encephalitis, Japanese/immunology , Encephalitis, Japanese/pathology , Encephalitis, Japanese/virology , Encephalitis, Viral/immunology , Hippocampus/pathology , Inflammation/pathology , Mice, Inbred ICR , Neurotoxins/toxicity , Receptors, CCR2/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
J Immunol Res ; 2021: 6654617, 2021.
Article in English | MEDLINE | ID: mdl-34041302

ABSTRACT

During the acute febrile phase of dengue virus (DENV) infection, viremia can cause severe systemic immune responses accompanied by hematologic disorders. This study investigated the potential induction and mechanism of the cytopathic effects of DENV on peripheral blood cells ex vivo. At one day postinfection, there was viral nonstructural protein NS1 but no further virus replication measured in the whole blood culture. Notably, DENV exposure caused significant vacuolization in monocytic phagocytes. With a minor change in the complete blood cell count, except for a minor increase in neutrophils and a significant decrease in monocytes, the immune profiling assay identified several changes, particularly a significant reduction in CD14-positive monocytes as well as CD11c-positive dendritic cells. Abnormal production of TNF-α was highly associated with the induction of vacuolization. Manipulating TNF-α expression resulted in cytopathogenic effects. These results demonstrate the potential hematological damage caused by ex vivo DENV-induced TNF-α.


Subject(s)
Dengue/immunology , Monocytes/pathology , Systemic Inflammatory Response Syndrome/immunology , Tumor Necrosis Factor-alpha/metabolism , Viremia/immunology , Aedes , Animals , Blood Cell Count , Cell Line , Coculture Techniques , Cricetinae , Dengue/blood , Dengue/complications , Dengue/virology , Dengue Virus/immunology , Healthy Volunteers , Humans , Monocytes/immunology , Primary Cell Culture , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/virology , Viremia/blood , Viremia/complications , Viremia/virology
11.
J Immunol Res ; 2021: 6680913, 2021.
Article in English | MEDLINE | ID: mdl-33869639

ABSTRACT

Propofol, 2,6-diisopropylphenol, is a short-acting intravenous sedative agent used in adults and children. Current studies show its various antimicrobial as well as anti-inflammatory effects. Dengue virus (DENV) is an emerging infectious pathogen transmitted by mosquitoes that causes mild dengue fever and progressive severe dengue diseases. In the absence of safe vaccines and antiviral agents, adjuvant treatments and supportive care are generally administered. This study investigated the antiviral effects of propofol against DENV infection and cellular inflammation by using an in vitro cell model. Treatment with propofol significantly inhibited DENV release 24 h postinfection in BHK-21 cells. Furthermore, it also blocked viral protein expression independent of the translational blockade. Propofol neither caused inhibitory effects on endosomal acidification nor prevented dsRNA replication. Either the proinflammatory TNF-α or the antiviral STAT1 signaling was reduced by propofol treatment. These results provide evidence to show the potential antiviral effects of the sedative propofol against DENV infection and cellular inflammation.


Subject(s)
Anesthetics/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/pharmacology , Dengue Virus/physiology , Dengue/drug therapy , Inflammation/drug therapy , Propofol/pharmacology , Animals , Cell Line , Humans , Immunity, Cellular , Interferon-gamma/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Virus Replication
12.
Front Cell Infect Microbiol ; 10: 557610, 2020.
Article in English | MEDLINE | ID: mdl-33072626

ABSTRACT

Dengue virus (DENV) infection in the brain causes severe dengue disease with neuropathic complications. In addition to viral effects, immunogenic or pathogenic central nervous system (CNS) inflammation can be induced during DENV infection. By using an immunocompetent outbred ICR (Institute of Cancer Research) mouse model for investigating CNS immunity upon DENV infection, we conducted single-panel immune cell profiling and a multiplex cytokine assay. The ICR mice infected with DENV presented with progressive hunchback posture, limbic seizures, limbic weakness, paralysis, and lethality. When the virions were released, the viral non-structural protein 1 was expressed in the brain in a time-dependent manner. Isolated brain CD45-positive cells revealed a significant population of resident CD14-positive cells, which was considerably decreased 8 days post-infection. We found an unexpected time-kinetic decrease in CD19-positive cells and CD11c/MHC II-positive cells and an increase in NK1.1-positive cells. Further assays showed the time-dependent induction of proinflammatory and NK1.1-associated cytokines in the DENV-infected brains. These results indicate a CNS immune profile of DENV infection and hypothetical CNS immunity in response to DENV infection.


Subject(s)
Dengue Virus , Dengue , Animals , Central Nervous System , Cytokines , Mice , Mice, Inbred ICR
13.
Article in English | MEDLINE | ID: mdl-32850477

ABSTRACT

Aging and chronic condition increase the incidence of dengue virus (DENV) infection, generally through a mechanism involving immunosenescence; however, the alternative effects of cellular senescence, which alters cell susceptibility to viral infection, remain unknown. Human monocytic THP-1 cells (ATCC TIB-202) treated with D-galactose to induce cellular senescence were susceptible to DENV infection. These senescent cells showed increased viral entry/binding, gene/protein expression, and dsRNA replication. The use of a replicon system showed that pharmacologically induced senescence did not enhance the effects on viral protein translation. By examining viral receptor expression, we found increased expression of CD209 (DC-SIGN) in the senescent cells. Interleukin (IL)-10 was aberrantly produced at high levels by the senescent cells, and the expression of the DENV receptor DC-SIGN was increased in these senescent cells, partially via IL-10-mediated regulation of the JAK2-STAT3 signaling pathway. The results demonstrate that a senescent phenotype facilitates DENV infection, probably by increasing DC-SIGN expression.


Subject(s)
Dengue Virus , Dengue , Humans , Monocytes , Virus Attachment , Virus Internalization , Virus Replication
14.
Front Cell Infect Microbiol ; 10: 606743, 2020.
Article in English | MEDLINE | ID: mdl-33634036

ABSTRACT

Dengue virus (DENV) is transmitted by Aedes mosquitoes to humans and is a threat worldwide. No effective new drugs have been used for anti-dengue treatment, and repurposing drugs is an alternative approach to treat this condition. Dopamine 2 receptor (D2R) is a host receptor positively associated with DENV infection. Metoclopramide (MCP), a D2R antagonist clinically used to control vomiting and nausea in patients with DENV infection, was putatively examined for inhibition of DENV infection by targeting D2R. In the mouse neural cell line Neuro-2a with D2R expression, a plaque assay demonstrated the antiviral efficacy of MCP treatment. However, in the cell line BHK-21, which did not express D2R, MCP treatment caused no further inhibition of DENV infection. Either MCP treatment or exogenous administration of a neutralizing D2R antibody blocked DENV binding. Treatment with MCP also reduced DENV dsRNA replication and DENV-induced neuronal cell cytotoxicity in vitro. An in vivo study demonstrated the antiviral effect of MCP against DENV-induced CNS neuropathy and mortality. These results showed that repurposing the D2R-targeting antiemetic MCP is a potential therapeutic strategy against DENV infection.


Subject(s)
Antiemetics , Dengue Virus , Dengue , Animals , Antiemetics/pharmacology , Antiemetics/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Dengue/drug therapy , Drug Repositioning , Humans , Metoclopramide/pharmacology , Metoclopramide/therapeutic use , Mice , Virus Replication
15.
J Immunol Res ; 2019: 1629258, 2019.
Article in English | MEDLINE | ID: mdl-31275997

ABSTRACT

The interferon- (IFN-) γ expression is elicited in response to microbial infections and activates immune surveillance by antimicrobial immune elements to induce microbial killing. Patients with adult-onset immunodeficiency who suffer from recurrent infections with microbes, particularly nontuberculous mycobacteria (NTM), commonly display genetic defects in IFN-γ signaling as well as the generation of anti-IFN-γ autoantibodies (autoAbs). Because IFN-γ is an activator of macrophage differentiation and a proinflammatory activator of innate immunity, the blockade effects of the autoAbs present in NTM patient serum on IFN-γ are hypothesized to regulate the antimicrobial function of macrophages. In the presence of patient serum, IFN-γ-induced type 1 macrophage (M1) differentiation was inhibited in PMA-stimulated human monocytic THP-1 cells. Treatment with patient serum significantly blocked the production of proinflammatory factors, including cytokines/chemokines and reactive oxygen/nitrogen species, by M1 macrophages. Importantly, IFN-γ-facilitated phagocytosis and degradation of heat-killed mycobacterium were decreased by cotreatment with patient serum. These results show the blockade activity of anti-IFN-γ autoAbs on IFN-γ-mediated antimicrobial immunity in macrophages.


Subject(s)
Antibodies, Blocking/immunology , Autoantibodies/immunology , Host-Pathogen Interactions/immunology , Immunomodulation , Interferon-gamma/immunology , Antibodies, Blocking/metabolism , Antibodies, Blocking/pharmacology , Autoantibodies/pharmacology , Biomarkers , Case-Control Studies , Cytokines/metabolism , Host-Pathogen Interactions/drug effects , Humans , Immunity, Innate , Interferon-gamma/antagonists & inhibitors , Macrophage Activation , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phagocytosis/immunology , Reactive Oxygen Species/metabolism , THP-1 Cells
16.
J Vis Exp ; (146)2019 04 28.
Article in English | MEDLINE | ID: mdl-31081826

ABSTRACT

Dengue virus (DENV), an arthropod-borne virus transmitted by mosquitoes, may cause the severe disease known as dengue hemorrhagic fever, which is characterized by lethal complications due to plasma leakage, ascites, pleural effusion, respiratory distress, severe bleeding, and organ impairment. A few cases of DENV infection present neurological manifestations; however, studies have not explored DENV-induced neuropathogenesis further. In this study, we present a protocol to use an immunocompetent outbred ICR (Institute of Cancer Research) mouse for investigating the induction of central nervous system (CNS) infection with DENV, followed by the progression of acute viral encephalitis-like disease.


Subject(s)
Dengue Virus/physiology , Disease Models, Animal , Encephalitis, Viral/virology , Acute Disease , Animals , Disease Progression , Mice , Mice, Inbred ICR
17.
Viruses ; 10(12)2018 12 05.
Article in English | MEDLINE | ID: mdl-30563082

ABSTRACT

The infection by dengue virus (DENV) of microglia causes cell activation and migration via a mechanism involving viral entry, RNA release, and Toll-like receptor 3 signaling. In this study, we demonstrated that secreted chemotactic factors present in microglial conditioned medium (MCM) facilitated cell motility in the murine BV2 microglial cells. The pharmacological disruption of lipid rafts/caveolae reduced DENV- and ultraviolet (UV)-inactivated MCM-induced microglial cell migration. An antibody-based cytokine/chemokine array showed an increase in macrophage inflammatory protein (MIP)-3ß in MCM produced using DENV-infected cells. The pharmacological inhibition of c-Jun N-terminal kinase (JNK) retarded UV-MCM-induced microglial cell migration. These results demonstrate that secreted MIP-3ß and its effect on the JNK signaling pathways mediates DENV-induced BV2 microglial cell migration.


Subject(s)
Cell Movement , Chemokine CCL19/immunology , Dengue Virus/immunology , Microglia/cytology , Signal Transduction , Animals , Caveolae/drug effects , Cell Line , Culture Media, Conditioned , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Membrane Microdomains/drug effects , Mice , Microglia/virology , Nystatin/pharmacology , Ultraviolet Rays
18.
PLoS Negl Trop Dis ; 12(8): e0006715, 2018 08.
Article in English | MEDLINE | ID: mdl-30125275

ABSTRACT

BACKGROUND: The antiparasitic agent niclosamide has been demonstrated to inhibit the arthropod-borne Zika virus. Here, we investigated the antiviral capacity of niclosamide against dengue virus (DENV) serotype 2 infection in vitro and in vivo. PRINCIPLE FINDING: Niclosamide effectively retarded DENV-induced infection in vitro in human adenocarcinoma cells (A549), mouse neuroblastoma cells (Neuro-2a), and baby hamster kidney fibroblasts (BHK-21). Treatment with niclosamide did not retard the endocytosis of DENV while niclosamide was unable to enhance the antiviral type I interferon response. Furthermore, niclosamide did not cause a direct effect on viral replicon-based expression. Niclosamide has been reported to competitively inhibit the mTOR (mammalian target of rapamycin), STAT3 (signal transducer and activator of transcription 3), and NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) signaling pathways; however, selective inhibitors of those pathways did not reduce DENV infection. Similar to the vacuolar-type H+-ATPase inhibitor bafilomycin A1, both niclosamide and other protonophores, such as CCCP (carbonyl cyanide m-chlorophenyl hydrazone), and FCCP (carbonyl cyanide-p-trifluoromethoxyphenylhydrazone), effectively reduced endosomal acidification and viral dsRNA replication. Co-administration of a single dose of niclosamide partially decreased viral replication, viral encephalitis, and mortality in DENV-infected ICR suckling mice. SIGNIFICANCE: These results demonstrate that niclosamide diminishes viral infection by hindering endosomal acidification.


Subject(s)
Dengue Virus/physiology , Endocytosis/drug effects , Endosomes/drug effects , Niclosamide/pharmacology , Animals , Animals, Suckling , Antiparasitic Agents , Antiviral Agents , Cell Line , Cricetinae , Endosomes/chemistry , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred ICR , Viral Plaque Assay
19.
J Leukoc Biol ; 104(5): 961-968, 2018 11.
Article in English | MEDLINE | ID: mdl-30044892

ABSTRACT

Proinflammatory TNF-α facilitates dengue virus (DENV) infection in endovascular dysfunction and neurotoxicity. The introduction of TNF-α blocking therapy with Abs is performed to test its therapeutic effect in this study. In DENV-infected mice, TNF-α production in the brain accompanied the progression of neurotoxicity and encephalitis. DENV infection caused the loss of hippocampal neurons with TNF-α expression around damaged regions, and immunostaining showed the induction of apoptosis in hippocampal neurons. TNF-α was expressed in active microglia and astrocytes in DENV-infected mice. TNF-α facilitated DENV-induced neurotoxicity in vitro in murine Neuro-2a cells. Using a currently established encephalitic mouse model in which DENV infection causes progressive hunchback posture, limbic seizures, limbic weakness, paralysis, and lethality 7 days postinfection, we showed that TNF-α transgenic mice represented the progressive disease development and administration of neutralizing TNF-α Ab reduced dengue encephalitis and mortality. These results demonstrate an immunopathogenesis of TNF-α for mediating DENV-induced encephalitis-associated neurotoxicity and that targeting TNF-α can be used as a strategy against dengue encephalitis.


Subject(s)
Dengue/immunology , Dengue/pathology , Encephalitis, Viral/immunology , Encephalitis, Viral/pathology , Tumor Necrosis Factor-alpha/immunology , Animals , Humans , Mice , Mice, Transgenic
20.
Virology ; 464-465: 218-227, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25104614

ABSTRACT

Enterovirus 71 (EV71) infection can induce encephalitis. Overt immune responses is suspected to cause severe symptoms, so anti-inflammatory agents, corticosteroids have been recommended for treatment. However, one clinical study reported that treatment with glucocorticoids, dexamethasone (Dex) exacerbates disease severity. Here we investigated Dex treatment on EV71 infection using the murine model and found that both long-term (14-day) and short-term (4-day) Dex treatment starting from 1 or 3 days postinfection increased the mortality and disease severity of infected mice. Dex treatment starting from 4 or 8 days postinfection did not affect mouse mortality and disease severity. Early Dex treatment starting from 1 day postinfection caused atrophy and enhanced apoptosis in lymphoid organs to decrease the numbers of lymphocytes (CD4(+) T cells, CD8(+) T cells, and CD19(+) B cells) and to increase viral loads in infected tissues of mice. Our results demonstrate that Dex treatment has no beneficial effect on EV71 infection.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Dexamethasone/adverse effects , Enterovirus A, Human/drug effects , Enterovirus Infections/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Apoptosis/drug effects , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Dexamethasone/administration & dosage , Enterovirus A, Human/physiology , Enterovirus Infections/mortality , Enterovirus Infections/physiopathology , Enterovirus Infections/virology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...