Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Immunol Immunother ; 73(10): 196, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105814

ABSTRACT

Anti-cluster of differentiation (CD) 3 × α programmed death-ligand 1 (PD-L1) bispecific T-cell engager (BsTE)-bound T-cells (BsTE:T) are a promising new cancer treatment agent. However, the mechanisms of action of bispecific antibody-armed activated T-cells are poorly understood. Therefore, this study aimed to investigate the anti-tumor mechanism and efficacy of BsTE:T. The BsTE:T migration was assessed in vivo and in vitro using syngeneic and xenogeneic tumor models, flow cytometry, immunofluorescence staining, transwell migration assays, microfluidic chips, Exo View R100, western blotting, and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology. In murine B16 melanoma, MC38 colon cancer, and human multiple myeloma cells, BsTE:T exhibited superior tumor elimination relative to that of T-cells or BsTE alone. Moreover, BsTE:T migration into tumors was significantly enhanced owing to the presence of PD-L1 in tumor cells and secretion of PD-L1-containing exosomes. Furthermore, increased infiltration of CD44highCD62Llow effector memory CD8+ T-cells into tumors was closely associated with the anti-tumor effect of BsTE:T. Therefore, BsTE:T is an innovative potential anti-tumor therapy, and exosomal PD-L1 plays a crucial role both in vitro and in vivo in the anti-tumor activity of BsTE:T.


Subject(s)
Antibodies, Bispecific , B7-H1 Antigen , Exosomes , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Exosomes/metabolism , Exosomes/immunology , Mice , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice, Inbred C57BL , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , CD3 Complex/immunology , CD3 Complex/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Cell Line, Tumor , Female , Cell Movement , Xenograft Model Antitumor Assays
2.
Mol Cancer ; 20(1): 133, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34649584

ABSTRACT

BACKGROUND: AMP-activated protein kinase (AMPK) is a metabolic sensor that maintains energy homeostasis. AMPK functions as a tumor suppressor in different cancers; however, its role in regulating antitumor immunity, particularly the function of regulatory T cells (Tregs), is poorly defined. METHODS: AMPKα1fl/flFoxp3YFP-Cre, Foxp3YFP-Cre, Rag1-/-, and C57BL/6 J mice were used for our research. Flow cytometry and cell sorting, western blotting, immuno-precipitation, immuno-fluorescence, glycolysis assay, and qRT-PCR were used to investigate the role of AMPK in suppressing programmed cell death 1 (PD-1) expression and for mechanistic investigation. RESULTS: The deletion of the AMPKα1 subunit in Tregs accelerates tumor growth by increasing the expression of PD-1. Metabolically, loss of AMPK in Tregs promotes glycolysis and the expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), a key enzyme of the mevalonate pathway. Mechanistically, AMPK activates the p38 mitogen-activated protein kinase (MAPK) that phosphorylates glycogen synthase kinase-3ß (GSK-3ß), inhibiting the expression of PD-1 in Tregs. CONCLUSION: Our study identified an AMPK regulatory mechanism of PD-1 expression via the HMGCR/p38 MAPK/GSK3ß signaling pathway. We propose that the AMPK activator can display synergic antitumor effect in murine tumor models, supporting their potential clinical use when combined with anti-PD-1 antibody, anti-CTLA-4 antibody, or a HMGCR inhibitor.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Immunomodulation , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Energy Metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Immunophenotyping , Mice , Programmed Cell Death 1 Receptor/metabolism
3.
Cytokine ; 127: 154983, 2020 03.
Article in English | MEDLINE | ID: mdl-31918161

ABSTRACT

Inflammasome activation induces the maturation and secretion of interleukin (IL)-1ß and -18, and is dependent on NF-κB signaling to induce the transcription of the inflammasome components, called the priming step. This study elucidated the role of IκBζ, an atypical IκBs (inhibitor of κB) and a coactivator of NF-κB target genes, on the activation of inflammasome. Bone marrow-derived macrophages (BMDMs) that originated from IκBζ-encoding Nfkbiz gene depletion mice presented a defect in NLRP3 inflammasome activation. In addition, the Nfkbiz+/- and Nfkbiz-/- mice significantly attenuated serum IL-1ß secretion in response to a monosodium urate injection, a NLRP3 trigger, when compared with Nfkbiz-+/+ mice. The lack of IκBζ in BMDMs produced a disability in the expression of Nlrp3 and pro-Il1ß mRNAs during the priming step. In addition, ectopic IκBζ expression enhanced the Nlrp3 promoter activity, and Nlrp3 and pro-Il1ß transcription. Overall, IκBζ controlled the activation of NLRP3 inflammasome by upregulating the Nlrp3 gene during the priming step.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Inflammasomes/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Up-Regulation/genetics , Animals , Cells, Cultured , Macrophages/metabolism , Mice , Promoter Regions, Genetic/genetics , RAW 264.7 Cells , RNA, Messenger/genetics , Signal Transduction/genetics , Transcription, Genetic/genetics
4.
J Cell Biochem ; 115(6): 1147-58, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24402912

ABSTRACT

Free fatty acid-induced lipotoxicity via increased endoplasmic reticulum (ER) stress and hepatocyte apoptosis is a key pathological mechanism of non-alcoholic steatohepatitis. A role of hypoxia-inducible factor 1α (HIF-1α) in this process has been suggested, but direct evidence is lacking. Here, we used HepG2 cells as a model to study whether HIF-1α can reduce palmitic acid-induced lipotoxicity and ER stress. In HepG2 cells treated with 500 µM palmitic acid, HIF-1α expression increased transiently, the decline was associated with increased cleaved caspase-3 expression. Overexpression and knockdown of HIF-1α decreased and exacerbated, respectively, palmitic acid-induced lipoapoptosis. The overexpression also blunted upregulation of the ER stress markers, C/EBP homologous protein (CHOP) and chaperone immunoglobulin heavy chain binding protein (Bip), while the knockdown increased the level of CHOP. In line with this, CHOP promoter activity decreased following HIF-1α binding to the CHOP promoter hypoxia response element. These results indicate that hepatocyte lipotoxicity is associated with decreased HIF-1α expression. It also suggests that upregulation of HIF-1α can be a possible strategy to reduce lipotoxicity in non-alcoholic fatty liver disease.


Subject(s)
Apoptosis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Palmitic Acid/pharmacology , Animals , Blotting, Western , Caspase 3/metabolism , Cell Hypoxia , Cell Survival/drug effects , Choline Deficiency , Diet , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver/metabolism , Liver/pathology , Methionine/deficiency , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protective Factors , Protein Binding , RNA Interference , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
5.
Biomol Ther (Seoul) ; 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39091181

ABSTRACT

ß-glucan, a polysaccharide found in various sources, exhibits unique physicochemical properties, yet its high polymerization limits clinical applications because of its solubility. Addressing this limitation, we introduce PPTEE-glycan, a highly purified soluble ß-1,3/1,6-glucan derived from Aureobasidium pullulans. The refined PPTEE-glycan demonstrated robust immune stimulation in vitro, activated dendritic cells, and enhanced co-stimulatory markers, cytokines, and cross-presentation. Formulated as a PPTEE + microemulsion (ME), it elevated immune responses in vivo, promoting antigen-specific antibodies and CD8+ T cell proliferation. Intratumoral administration of PPTEE + ME in tumor-bearing mice induced notable tumor regression, which was linked to the activation of immunosuppressive cells. This study highlights the potential of high-purity Aureobasidium pullulans-derived ß-glucan, particularly PPTEE, as promising immune adjuvants, offering novel avenues for advancing cancer immunotherapy.

6.
J Control Release ; 366: 142-159, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38145660

ABSTRACT

Responsive heat resistance (by heat shock protein upregulation) and spontaneous reactive oxygen species (ROS) detoxification have been regarded as the major obstacles for photothermal/photodynamic therapy of cancer. To overcome the thermal resistance and improve ROS susceptibility in breast cancer therapy, Au ion-crosslinked hydrogels including indocyanine green (ICG) and polyphenol are devised. Au ion has been introduced for gel crosslinking (by catechol-Au3+ coordination), cellular glutathione depletion, and O2 production from cellular H2O2. ICG can generate singlet oxygen from O2 (for photodynamic therapy) and induce hyperthermia (for photothermal therapy) under the near-infrared laser exposure. (-)-Epigallocatechin gallate downregulates heat shock protein to overcome heat resistance during hyperthermia and exerts multiple anticancer functions in spite of its ironical antioxidant features. Those molecules are concinnously engaged in the hydrogel structure to offer fast gel transformation, syringe injection, self-restoration, and rheological tuning for augmented photo/chemotherapy of cancer. Intratumoral injection of multifunctional hydrogel efficiently suppressed the growth of primary breast cancer and completely eliminated the residual tumor mass. Proposed hydrogel system can be applied to tumor size reduction prior to surgery of breast cancer and the complete remission after its surgery.


Subject(s)
Breast Neoplasms , Hyperthermia, Induced , Photochemotherapy , Humans , Female , Reactive Oxygen Species/metabolism , Hydrogels/therapeutic use , Hydrogen Peroxide , Indocyanine Green/therapeutic use , Indocyanine Green/chemistry , Breast Neoplasms/drug therapy , Heat-Shock Proteins
7.
J Control Release ; 362: 1-18, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37595669

ABSTRACT

Alum-crosslinked hyaluronic acid-dopamine (HD) hydrogel containing indocyanine green (ICG) with anti-programmed cell death-1 (PD-1) antibody (Ab) administration was developed for immunophoto therapy of cancer. Alum modulates the rheological characteristics of hydrogel for enabling syringe injection, shear-thinning feature, and slower biodegradation. In addition, alum in HD-based hydrogel provided CD8+ T cell-mediated immune responses for cancer therapy. ICG in the hydrogel under near-infrared (NIR) light exposure may induce hyperthermia and generate singlet oxygen for selective cancer cell killing. HD/alum/ICG hydrogel injection with NIR laser irradiation elevated PD-1 level in CD8+ T cells. Administration of PD-1 Ab aiming at highly expressed PD-1 in T cells may amplify the anticancer efficacies of HD/alum/ICG hydrogel along with NIR laser. HD/alum/ICG hydrogel with NIR light may have both CD8+ T cell-linked immune responses and ICG-related photodynamic/photothermal effects. Additional injection of immune checkpoint inhibitor can ultimately suppress primary and distant tumor growth by combination with those therapeutic actions.

8.
Nat Commun ; 14(1): 2593, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37147330

ABSTRACT

Programmed cell death protein 1 (PD-1), expressed on tumor-infiltrating T cells, is a T cell exhaustion marker. The mechanisms underlying PD-1 upregulation in CD4 T cells remain unknown. Here we develop nutrient-deprived media and a conditional knockout female mouse model to study the mechanism underlying PD-1 upregulation. Reduced methionine increases PD-1 expression on CD4 T cells. The genetic ablation of SLC43A2 in cancer cells restores methionine metabolism in CD4 T cells, increasing the intracellular levels of S-adenosylmethionine and yielding H3K79me2. Reduced H3K79me2 due to methionine deprivation downregulates AMPK, upregulates PD-1 expression and impairs antitumor immunity in CD4 T cells. Methionine supplementation restores H3K79 methylation and AMPK expression, lowering PD-1 levels. AMPK-deficient CD4 T cells exhibit increased endoplasmic reticulum stress and Xbp1s transcript levels. Our results demonstrate that AMPK is a methionine-dependent regulator of the epigenetic control of PD-1 expression in CD4 T cells, a metabolic checkpoint for CD4 T cell exhaustion.


Subject(s)
CD4-Positive T-Lymphocytes , Neoplasms , Programmed Cell Death 1 Receptor , Animals , Female , Mice , AMP-Activated Protein Kinases/metabolism , CD8-Positive T-Lymphocytes , Methionine/metabolism , Mice, Knockout , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Racemethionine/metabolism , Up-Regulation
9.
Exp Mol Med ; 55(2): 401-412, 2023 02.
Article in English | MEDLINE | ID: mdl-36759578

ABSTRACT

Endoplasmic reticulum stress is closely associated with the onset and progression of inflammatory bowel disease. ERdj5 is an endoplasmic reticulum-resident protein disulfide reductase that mediates the cleavage and degradation of misfolded proteins. Although ERdj5 expression is significantly higher in the colonic tissues of patients with inflammatory bowel disease than in healthy controls, its role in inflammatory bowel disease has not yet been reported. In the current study, we used ERdj5-knockout mice to investigate the potential roles of ERdj5 in inflammatory bowel disease. ERdj5 deficiency causes severe inflammation in mouse colitis models and weakens gut barrier function by increasing NF-κB-mediated inflammation. ERdj5 may not be indispensable for goblet cell function under steady-state conditions, but its deficiency induces goblet cell apoptosis under inflammatory conditions. Treatment of ERdj5-knockout mice with the chemical chaperone ursodeoxycholic acid ameliorated severe colitis by reducing endoplasmic reticulum stress. These findings highlight the important role of ERdj5 in preserving goblet cell viability and function by resolving endoplasmic reticulum stress.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Mice , HSP40 Heat-Shock Proteins/metabolism , Protein Folding , Goblet Cells/metabolism , Inflammation , Mice, Knockout , Endoplasmic Reticulum Stress , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Apoptosis , Molecular Chaperones/metabolism
10.
Biomol Ther (Seoul) ; 30(5): 418-426, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35577765

ABSTRACT

Chimeric antigen receptor T (CAR-T) cell therapy is one of the promising anticancer treatments. It shows a high overall response rate with complete response to blood cancer. However, there is a limitation to solid tumor treatment. Additionally, this currently approved therapy exhibits side effects such as cytokine release syndrome and neurotoxicity. Alternatively, bispecific antibody is an innovative therapeutic tool that simultaneously engages specific immune cells to disease-related target cells. Since programmed death ligand 1 (PD-L1) is an immune checkpoint molecule highly expressed in some cancer cells, in the current study, we generated αCD3xαPD-L1 bispecific antibody (BiTE) which can engage T cells to PD-L1+ cancer cells. We observed that the BiTE-bound OT-1 T cells effectively killed cancer cells in vitro and in vivo. They substantially increased the recruitment of effector memory CD8+ T cells having CD8+CD44+CD62Llow phenotype in tumor. Interestingly, we also observed that BiTE-bound polyclonal T cells showed highly efficacious tumor killing activity in vivo in comparison with the direct intravenous treatment of bispecific antibody, suggesting that PD-L1-directed migration and engagement of activated T cells might increase cancer cell killing. Additionally, BiTE-bound CAR-T cells which targets human Her-2/neu exhibited enhanced killing effect on Her-2-expressing cancer cells in vivo, suggesting that this could be a novel therapeutic regimen. Collectively, our results suggested that engaging activated T cells with cancer cells using αCD3xαPD-L1 BiTE could be an innovative next generation anticancer therapy which exerts simultaneous inhibitory functions on PD-L1 as well as increasing the infiltration of activated T cells having effector memory phenotype in tumor site.

11.
Immune Netw ; 22(2): e19, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35573153

ABSTRACT

Coxsackievirus B3 (CVB3) infection causes acute pancreatitis and myocarditis. However, its pathophysiological mechanism is unclear. Here, we investigated how lipid metabolism is associated with exacerbation of CVB3 pathology using high-fat diet (HFD)-induced obese mice. Mice were intraperitoneally inoculated with 1×106 pfu/mouse of CVB3 after being fed a control or HFD to induce obesity. Mice were treated with mitoquinone (MitoQ) to reduce the level of mitochondrial ROS (mtROS). In obese mice, lipotoxicity of white adipose tissue-induced inflammation caused increased replication of CVB3 and mortality. The coxsackievirus adenovirus receptor increased under obese conditions, facilitating CVB3 replication in vitro. However, lipid-treated cells with receptor-specific inhibitors did not reduce CVB3 replication. In addition, lipid treatment increased mitochondria-derived vesicle formation and the number of multivesicular bodies. Alternatively, we found that inhibition of lipid-induced mtROS decreased viral replication. Notably, HFD-fed mice were more susceptible to CVB3-induced mortality in association with increased levels of CVB3 replication in adipose tissue, which was ameliorated by administration of the mtROS inhibitor, MitoQ. These results suggest that mtROS inhibitors can be used as potential treatments for CVB3 infection.

12.
Free Radic Biol Med ; 184: 42-52, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35390453

ABSTRACT

Alcoholic liver disease is the major cause of chronic liver diseases. Excessive alcohol intake results in endoplasmic reticulum (ER) stress. ERdj5, a member of DNAJ family, is an ER-resident chaperone protein, whose role in alcoholic liver disease remains to be investigated. In this study, we aim to address the effect of ERdj5 on alcoholic liver disease and the underlying mechanism. Hepatic Dnajc10 (ERdj5) mRNA expression was elevated in both human and mouse alcoholic hepatitis. In mice subjected to chronic and binge ethanol feeding, ERdj5 levels were also markedly increased. Hepatic Dnajc10 correlated with Xbp1s mRNA. Tunicamycin, an ER stress inducer, increased ERdj5 levels. Dnajc10 knockout mice exhibited exacerbated alcohol-induced liver injury and hepatic steatosis. However, the macrophage numbers and chemokine levels were similar to those in wild-type mice. Depletion of Dnajc10 promoted oxidative stress. Ethanol feeding increased hepatic H2O2 levels, and these were further increased in Dnajc10 knockout mice. Additionally, Dnajc10-deficient hepatocytes produced large amounts of reactive oxygen species. Notably, Nrf2, a central regulator of oxidative stress, was decreased by depletion of Dnajc10 in the nuclear fraction of ethanol-treated mouse liver. Consistently, liver tissues from ethanol-fed Dnajc10 knockout mice had reduced expression of downstream antioxidant genes. Furthermore, hepatic glutathione content in the liver of knockout mice declined compared to wild-type mice. In conclusion, our results demonstrate that ethanol-induced ERdj5 may regulate the Nrf2 pathway and glutathione contents, and have protective effects on liver damage and alcohol-mediated oxidative stress in mice. These suggest that ERdj5 has the potential to protect against alcoholic liver disease.


Subject(s)
HSP40 Heat-Shock Proteins , Liver Diseases, Alcoholic , Molecular Chaperones , NF-E2-Related Factor 2 , Animals , Mice , Ethanol/toxicity , Glutathione/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Hydrogen Peroxide/metabolism , Liver/metabolism , Liver Diseases, Alcoholic/genetics , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , RNA, Messenger/metabolism
13.
Biomol Ther (Seoul) ; 29(2): 166-174, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33139584

ABSTRACT

Multiple myeloma is a malignant cancer of plasma cells. Despite recent progress with immunomodulatory drugs and proteasome inhibitors, it remains an incurable disease that requires other strategies to overcome its recurrence and non-response. Based on the high expression levels of programmed death-ligand 1 (PD-L1) in human multiple myeloma isolated from bone marrow and the murine myeloma cell lines, NS-1 and MOPC-315, we propose PD-L1 molecule as a target of anti-multiple myeloma therapy. We developed a novel anti-PD-L1 antibody containing a murine immunoglobulin G subclass 2a (IgG2a) fragment crystallizable (Fc) domain that can induce antibody-dependent cellular cytotoxicity. The newly developed anti-PD-L1 antibody showed significant antitumor effects against multiple myeloma in mice subcutaneously, intraperitoneally, or intravenously inoculated with NS-1 and MOPC-315 cells. The anti-PD-L1 effects on multiple myeloma may be related to a decrease in the immunosuppressive myeloid-derived suppressor cells (MDSCs), but there were no changes in the splenic MDSCs after combined treatment with lenalidomide and the anti-PD-L1 antibody. Interestingly, the newly developed anti-PD-L1 antibody can induce antibody-dependent cellular cytotoxicity in the myeloma cells, which differs from the existing anti-PD-L1 antibodies. Collectively, we have developed a new anti-PD-L1 antibody that binds to mouse and human PD-L1 and demonstrated the antitumor effects of the antibody in several syngeneic murine myeloma models. Thus, PD-L1 is a promising target to treat multiple myeloma, and the novel anti-PD-L1 antibody may be an effective anti-myeloma drug via antibody-dependent cellular cytotoxicity effects.

14.
Mol Cells ; 43(12): 989-1001, 2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33250450

ABSTRACT

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a facultative intracellular pathogen that causes salmonellosis and mortality worldwide. S. Typhimurium infects macrophages and survives within phagosomes by avoiding the phagosome-lysosome fusion system. Phagosomes sequentially acquire different Rab GTPases during maturation and eventually fuse with acidic lysosomes. Lysophosphatidylcholine (LPC) is a bioactive lipid that is associated with the generation of chemoattractants and reactive oxygen species (ROS). In our previous study, LPC controlled the intracellular growth of Mycobacterium tuberculosis by promoting phagosome maturation. In this study, to verify whether LPC enhances phagosome maturation and regulates the intracellular growth of S. Typhimurium, macrophages were infected with S. Typhimurium. LPC decreased the intracellular bacterial burden, but it did not induce cytotoxicity in S. Typhimuriuminfected cells. In addition, combined administration of LPC and antibiotic significantly reduced the bacterial burden in the spleen and the liver. The ratios of the colocalization of intracellular S. Typhimurium with phagosome maturation markers, such as early endosome antigen 1 (EEA1) and lysosome-associated membrane protein 1 (LAMP-1), were significantly increased in LPC-treated cells. The expression level of cleaved cathepsin D was rapidly increased in LPCtreated cells during S. Typhimurium infection. Treatment with LPC enhanced ROS production, but it did not affect nitric oxide production in S. Typhimurium-infected cells. LPC also rapidly triggered the phosphorylation of IκBα during S. Typhimurium infection. These results suggest that LPC can improve phagosome maturation via ROS-induced activation of NF-κB pathway and thus may be developed as a therapeutic agent to control S. Typhimurium growth.


Subject(s)
Lysophosphatidylcholines/metabolism , Macrophages/metabolism , Macrophages/microbiology , NF-kappa B/metabolism , Phagosomes/metabolism , Salmonella Infections/metabolism , Signal Transduction , Animals , Male , Mice , Mice, Inbred BALB C , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/physiology
15.
Immune Netw ; 20(4): e32, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32895619

ABSTRACT

Influenza virus is the major cause of seasonal and pandemic flu. Currently, oseltamivir, a potent and selective inhibitor of neuraminidase of influenza A and B viruses, is the drug of choice for treating patients with influenza virus infection. However, recent emergence of oseltamivir-resistant influenza viruses has limited its efficacy. Morin hydrate (3,5,7,2',4'-pentahydroxyflavone) is a flavonoid isolated from Morus alba L. It has antioxidant, anti-inflammatory, neuroprotective, and anticancer effects partly by the inhibition of the NF-кB signaling pathway. However, its effects on influenza virus have not been studied. We evaluated the antiviral activity of morin hydrate against influenza A/Puerto Rico/8/1934 (A/PR/8; H1N1) and oseltamivir-resistant A/PR/8 influenza viruses in vitro. To determine its mode of action, we carried out time course experiments, and time of addition, hemolysis inhibition, and hemagglutination assays. The effects of the co-administration of morin hydrate and oseltamivir were assessed using the murine model of A/PR/8 infection. We found that morin hydrate reduced hemagglutination by A/PR/8 in vitro. It alleviated the symptoms of A/PR/8-infection, and reduced the levels of pro-inflammatory cytokines and chemokines, such as TNF-α and CCL2, in infected mice. Co-administration of morin hydrate and oseltamivir phosphate reduced the virus titers and attenuated pulmonary inflammation. Our results suggest that morin hydrate exhibits antiviral activity by inhibiting the entry of the virus.

16.
Cancer Lett ; 493: 102-112, 2020 11 28.
Article in English | MEDLINE | ID: mdl-32810576

ABSTRACT

Toll-like receptor (TLR)3 and TLR7 are important for stimulating plasmacytoid dendritic cells (pDCs), which secrete type I interferon. Mice deficient for TLR3 and TLR7 (TLR3-/-TLR7-/-) reportedly exhibit deteriorated colitis because of impaired pDCs. However, the role of pDCs in tumorigenesis-associated inflammation progression has not been studied. We treated wild-type or TLR3-/-TLR7-/- mice with dextran sulfate sodium (DSS) and/or azoxymethane (AOM) and examined colon mucosa, measured body weight and colon length of mice, and examined pDC and myeloid-derived suppressor cell (MDSC) accumulation. Further, we depleted pDCs in AOM/DSS-treated wild-type mice by treating them with anti-PDCA-1 antibodies. We found that MDSCs significantly increased, while pDCs decreased in TLR3-/-TLR7-/- mice. Moreover, TLR3-/-TLR7-/- mice developed colitis-associated colon cancer following AOM/DSS treatment. Additionally, we showed that a defect in TLR7 of pDCs is responsible for the aggravation of colitis-associated colon cancer. Further, we showed that TLR7 ligand mitigates colitis-associated colon cancer. Collectively, our results demonstrate that gut pDCs play a crucial role in reducing colorectal cancer development via the regulation of infiltrating MDSCs.


Subject(s)
Colitis/complications , Colonic Neoplasms/pathology , Dendritic Cells/metabolism , Membrane Glycoproteins/genetics , Myeloid-Derived Suppressor Cells/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 7/genetics , Animals , Azoxymethane/adverse effects , Body Weight , Cell Line, Tumor , Colitis/chemically induced , Colitis/genetics , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Disease Progression , Female , Gene Knockout Techniques , Mice , Signal Transduction
17.
Toxicol Appl Pharmacol ; 239(1): 71-9, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19467251

ABSTRACT

Dithiocarbamates are a commercially important class of compounds that can produce peripheral neuropathy in humans and experimental animals. Previous studies have supported a requirement for copper accumulation and enhanced lipid peroxidation in dithiocarbamate-mediated myelinopathy. The study presented here extends previous investigations in two areas. Firstly, although total copper levels have been shown to increase within the nerve it has not been determined whether copper is increased within the myelin compartment, the primary site of lesion development. Therefore, the distribution of copper in sciatic nerve was characterized using synchrotron X-ray fluorescence microscopy to determine whether the neurotoxic dithiocarbamate, N,N-diethyldithiocarbamate, increases copper levels in myelin. Secondly, because lipid peroxidation is an ongoing process in normal nerve and the levels of lipid peroxidation products produced by dithiocarbamate exposure demonstrated an unusual cumulative dose response in previous studies the biological impact of dithiocarbamate-mediated lipid peroxidation was evaluated. Experiments were performed to determine whether dithiocarbamate-mediated lipid peroxidation products elicit an antioxidant response through measuring the protein expression levels of three enzymes, superoxide dismutase 1, heme oxygenase 1, and glutathione transferase alpha, that are linked to the antioxidant response element promoter. To establish the potential of oxidative injury to contribute to myelin injury the temporal relationship of the antioxidant response to myelin injury was determined. Myelin structure in peripheral nerve was assessed using multi-exponential transverse relaxation measurements (MET(2)) as a function of exposure duration, and the temporal relationship of protein expression changes relative to the onset of changes in myelin integrity were determined. Initial assessments were also performed to explore the potential contribution of dithiocarbamate-mediated inhibition of proteasome function and inhibition of cuproenzyme activity to neurotoxicity, and also to assess the potential of dithiocarbamates to promote oxidative stress and injury within the central nervous system. These evaluations were performed using an established model for dithiocarbamate-mediated demyelination in the rat utilizing sciatic nerve, spinal cord and brain samples obtained from rats exposed to N,N-diethyldithiocarbamate (DEDC) by intra-abdominal pumps for periods of 2, 4, and 8 weeks and from non exposed controls. The data supported the ability of DEDC to increase copper within myelin and to enhance oxidative stress prior to structural changes detectable by MET(2). Evidence was also obtained that the excess copper produced by DEDC in the central nervous system is redox active and promotes oxidative injury.


Subject(s)
Copper/metabolism , Ditiocarb/toxicity , Myelin Sheath/drug effects , Oxidative Stress/drug effects , Animals , Blotting, Western , Brain/drug effects , Brain/enzymology , Brain/metabolism , Brain/ultrastructure , Glutathione Transferase/biosynthesis , Heme Oxygenase (Decyclizing)/biosynthesis , Isoenzymes/biosynthesis , Lipid Peroxidation/drug effects , Male , Microscopy, Fluorescence , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Proteasome Endopeptidase Complex/metabolism , Protein Carbonylation , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/enzymology , Sciatic Nerve/metabolism , Sciatic Nerve/ultrastructure , Spinal Cord/drug effects , Spinal Cord/enzymology , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Superoxide Dismutase/biosynthesis , Superoxide Dismutase-1
19.
Front Immunol ; 10: 2542, 2019.
Article in English | MEDLINE | ID: mdl-31736965

ABSTRACT

Early secretory antigenic target-6 (ESAT6) is a potent immunogenic antigen expressed in Mycobacterium tuberculosis as well as in some non-tuberculous mycobacteria (NTM), such as M. kansasii. M. kansasii is one of the most clinically relevant species of NTM that causes mycobacterial lung disease, which is clinically indistinguishable from tuberculosis. In the current study, we designed a novel cell-based vaccine using B cells that were transduced with vaccinia virus expressing ESAT6 (vacESAT6), and presenting α-galactosylceramide (αGC), a ligand of invariant NKT cells. We found that B cells loaded with αGC had increased levels of CD80 and CD86 after in vitro stimulation with NKT cells. Immunization of mice with B/αGC/vacESAT6 induced CD4+ T cells producing TNF-α and IFN-γ in response to heat-killed M. tuberculosis. Immunization of mice with B/αGC/vacESAT6 ameliorated severe lung inflammation caused by M. kansasii infection. We also confirmed that immunization with B/αGC/vacESAT6 reduced M. kansasii bacterial burden in the lungs. In addition, therapeutic administration of B/αGC/vacESAT6 increased IFN-γ+ CD4+ T cells and inhibited the progression of lung pathology caused by M. kansasii infection. Thus, B/αGC/vacESAT6 could be a potent vaccine candidate for the prevention and treatment of ESAT6-expressing mycobacterial infection caused by M. kansasii.


Subject(s)
Antigens, Bacterial/immunology , B-Lymphocytes/immunology , Bacterial Proteins/immunology , Galactosylceramides/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Vaccinia virus , Animals , Antibodies, Bacterial/immunology , Antigen Presentation/immunology , Antigens, Bacterial/genetics , B-Lymphocytes/metabolism , Bacterial Proteins/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Immunization , Mice , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Vaccinia virus/genetics , Vaccinia virus/immunology
20.
Sci Rep ; 9(1): 8397, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182790

ABSTRACT

Inhibitor of kappa B (IκB)-ζ transcription is rapidly induced by stimulation with TLR ligands and IL-1. Despite high IκBζ expression in inflammation sites, the association of IκBζ with host defence via systemic immune responses against bacterial infection remains unclear. Oral immunisation with a recombinant attenuated Salmonella vaccine (RASV) strain did not protect IκBζ-deficient mice against a lethal Salmonella challenge. IκBζ-deficient mice failed to produce Salmonella LPS-specific IgG, especially IgG2a, although inflammatory cytokine production and immune cell infiltration into the liver increased after oral RASV administration. Moreover, IκBζ-deficient mice exhibited enhanced splenic germinal centre reactions followed by increased total IgG production, despite IκBζ-deficient B cells having an intrinsic antibody class switching defect. IκBζ-deficient CD4+ T cells poorly differentiated into Th1 cells. IFN-γ production by CD4+ T cells from IκBζ-deficient mice immunised with RASV significantly decreased after restimulation with heat-killed RASV in vitro, suggesting that IκBζ-deficient mice failed to mount protective immune responses against Salmonella infection because of insufficient Th1 and IgG production. Therefore, IκBζ is crucial in protecting against Salmonella infection by inducing Th1 differentiation followed by IgG production.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Immunity , Immunoglobulin G/biosynthesis , Salmonella Infections/immunology , Salmonella Infections/prevention & control , Th1 Cells/immunology , Administration, Oral , Animals , Chronic Disease , Germinal Center/metabolism , Immunization , Inflammation/pathology , Interferon-gamma/metabolism , Lipopolysaccharides , Mice, Inbred C57BL , Salmonella Infections/parasitology , Salmonella Vaccines/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Vaccines, Attenuated/immunology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL