Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Innate Immun ; 24(5): 323-331, 2018 07.
Article in English | MEDLINE | ID: mdl-29848176

ABSTRACT

The TLR2 agonist, dipalmitoyl lipopeptide (Pam2LP), has been used as an immune adjuvant without much success. Pam2LP is recognised by TLR2/6 receptors in humans and in mice. This study examined the proliferative activity of cytotoxic T lymphocytes (CTL) using mouse Ag-presenting dendritic cells (DCs) and OT-I assay system, where a library of synthetic Pam2LP was utilised from the Staphylococcus aureus database. Ag-specific CTL expansion and IFN-γ levels largely depended on the Pam2LP peptide sequence. The first Aa is cysteine (Cys), which has an active SH residue to bridge fatty acids, and the second and third Aa are hydrophilic or non-polar. The sequence structurally adapted to the residual constitution of the reported TLR2/6 pocket. The inactive sequence contained proline or leucine/isoleucine after the first Cys. Notably, no direct activation of OT-I cells was detected without DCs by stimulation with the active Pam2LP having the Cys-Ser sequence. MyD88, but not TICAM-1 or IFN pathways, in DCs participates in DC maturation characterised by upregulation of CD40, CD80 and CD86. Hence, the active Pam2LPs appear suitable for dimeric TLR2/6 on DCs, resulting in induction of DC maturation.


Subject(s)
Cysteine/metabolism , Dendritic Cells/immunology , Lipopeptides/metabolism , Serine/metabolism , Staphylococcus aureus/physiology , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic , Animals , Antigen Presentation , Cell Differentiation , Cell Proliferation , Cells, Cultured , Interferon-gamma/metabolism , Lipopeptides/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Peptide Library , Protein Conformation , Staphylococcus aureus/metabolism , Toll-Like Receptor 2/agonists
2.
Proc Jpn Acad Ser B Phys Biol Sci ; 94(3): 153-160, 2018.
Article in English | MEDLINE | ID: mdl-29526974

ABSTRACT

The immune system eliminates advanced cancer when treated with programmed cell death protein-1 (PD-1) or its ligand (PD-L1) blockade, but PD-1 therapy is effective in only ∼20% of patients with solid cancer. The PD-1 antibody mainly acts on the effector phase of cytotoxic T lymphocytes (CTLs) in tumors but induces no activation of the priming phase of antigen-presenting dendritic cells (DCs). It is reasonable that both DC-priming and PD-1/L1 blocking are mandatory for efficient CTL-mediated tumor cytolysis. For DC-priming, a therapeutic vaccine containing Toll-like receptor (TLR) agonists, namely a priming adjuvant, is a good candidate; however, a means for DC-targeting by TLR adjuvant therapy remains to be developed. TLR adjuvants usually harbor cytokine toxicity, which is a substantial barrier against drug approval. Here, we discuss the functional properties of current TLR adjuvants for cancer immunotherapy and introduce a TLR3-specific adjuvant (ARNAX) that barely induces cytokinemia in mouse models.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans
3.
Front Immunol ; 9: 496, 2018.
Article in English | MEDLINE | ID: mdl-29593736

ABSTRACT

Mycoplasma fermentans-derived diacylated lipoprotein M161Ag (MALP404) is recognized by human/mouse toll-like receptor (TLR) 2/TLR6. Short proteolytic products including macrophage-activating lipopeptide 2 (MALP2) have been utilized as antitumor immune-enhancing adjuvants. We have chemically synthesized a short form of MALP2 named MALP2s (S-[2,3-bis(palmitoyloxy)propyl]-CGNNDE). MALP2 and MALP2s provoke natural killer (NK) cell activation in vitro but only poorly induce tumor regression using in vivo mouse models loading NK-sensitive tumors. Here, we identified the functional mechanism of MALP2s on dendritic cell (DC)-priming and cytotoxic T lymphocyte (CTL)-dependent tumor eradication using CTL-sensitive tumor-implant models EG7 and B16-OVA. Programmed death ligand-1 (PD-L1) blockade therapy in combination with MALP2s + ovalbumin (OVA) showed a significant additive effect on tumor growth suppression. MALP2s increased co-stimulators CD80/86 and CD40, which were totally MyD88-dependent, with no participation of toll-IL-1R homology domain-containing adaptor molecule-1 or type I interferon signaling in DC priming. MALP2s + OVA consequently augmented proliferation of OVA-specific CTLs in the spleen and at tumor sites. Chemokines and cytolytic factors were upregulated in the tumor. Strikingly, longer duration and reinvigoration of CTLs in spleen and tumors were accomplished by the addition of MALP2s + OVA to α-PD-L1 antibody (Ab) therapy compared to α-PD-L1 Ab monotherapy. Then, tumors regressed better in the MALP2s/OVA combination than in the α-PD-L1 Ab monotherapy. Hence, MALP2s/tumor-associated antigens combined with α-PD-L1 Ab is a good therapeutic strategy in some mouse models. Unfortunately, numerous patients are still resistant to PD-1/PD-L1 blockade, and good DC-priming adjuvants are desired. Cytokine toxicity by MALP2s remains to be settled, which should be improved by chemical modification in future studies.


Subject(s)
Bacterial Proteins/pharmacology , Dendritic Cells/immunology , Lipopeptides/pharmacology , Lymphocyte Activation/drug effects , Mycoplasma fermentans/chemistry , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Bacterial Proteins/chemistry , Cell Line, Tumor , Dendritic Cells/pathology , Interferon Type I/genetics , Interferon Type I/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lipopeptides/chemistry , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , T-Lymphocytes, Cytotoxic/pathology
4.
Oncoimmunology ; 5(8): e1188244, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27622060

ABSTRACT

Successful cancer immunotherapy necessitates T cell proliferation and infiltration into tumor without exhaustion, a process closely links optimal maturation of dendritic cells (DC), and adjuvant promotes this process as an essential prerequisite. Poly(I:C) has contributed to adjuvant immunotherapy that evokes an antitumor response through the Toll-loke receptor 3 (TLR3)/TICAM-1 pathway in DC. However, the mechanism whereby Poly(I:C) acts on DC for T cell proliferation and migration remains undetermined. Subcutaneous injection of Poly(I:C) regressed implant tumors (WT1-C1498 or OVA-EG7) in C57BL/6 mice, which coincided with tumor-infiltration of CD8(+) T cells. Epitope-specific cytotoxic T lymphocytes (CTLs) were increased in spleen by challenge with Poly(I:C)+Db126 WT-1 peptide but not Poly(I:C) alone, suggesting the need of an exogenous Ag density for cross-priming. In tumor, CXCR3 ligands were upregulated by Poly(I:C), which facilitated recruitment of CTL to the tumor. Thus, Poly(I:C) acts on splenic CD8α(+) DC to cross-prime T cells and on intratumor cells to attract CTLs. Besides CD8(+) T cell cross-priming, T cell recruitment into tumor was significantly dampened in Batf3 (-/-) mice, reflecting the importance of tumor Batf3-dependent DC rather than macrophages in T cell recruitment. Poly(I:C)-induced XCR1(hi) CD8α(+) DC with high TLR3 levels were markedly decreased in Batf3 (-/-) mice, which hampered the production of IL-12 and IL-12-mediated CD4(+)/CD8(+) T cell proliferation. Subcutaneous administration of Poly(I:C) and adoptive transfer of wild-type CD8α(+) DC largely recovered antitumor response in those Batf3 (-/-) mice. Collectively, Poly(I:C) tunes up proper maturation of CD8α(+) DC to establish TLR3-mediated IL-12 function and cross-presentation in spleen and lymphocyte-attractive antitumor microenvironment in tumor.

5.
J Exp Clin Cancer Res ; 35(1): 143, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27619885

ABSTRACT

BACKGROUND: Dendritic cells (DCs) mount tumor-associated antigens (TAAs), and the double-stranded RNA adjuvant Poly(I:C) stimulates Toll-like receptor 3 (TLR3) signal in DC, which in turn induces type I interferon (IFN) and interleukin-12 (IL-12), then cross-primes cytotoxic T lymphocytes (CTLs). Proliferation of CTLs correlates with tumor regression. How these potent cells expand with high quality is crucial to the outcome of CTL therapy. However, good markers reflecting the efficacy of DC-target immunotherapy have not been addressed. METHODS: Using an EG7 (ovalbumin, OVA-positive) tumor-implant mouse model, we examined what is a good marker for active CTL induction in treatment with Poly(I:C)/OVA. RESULTS: Simultaneous administration of Poly(I:C) and antigen (Ag) OVA significantly increased a minor population of CD8(+) T cells, that express CD11c in lymphoid and tumor sites. The numbers of the CD11c(+) CD8(+) T cells correlated with those of induced Ag-specific CD8(+) T cells and tumor regression. The CD11c(+) CD8(+) T cell moiety was characterized by its high killing activity and IFN-γ-producing ability, which represent an active phenotype of the effector CTLs. Not only a TLR3-specific (TICAM-1-dependent) signal but also TLR2 (MyD88) signal in DC triggered the expansion of CD11c(+) CD8(+) T cells in tumor-bearing mice. Notably, human CD11c(+) CD8(+) T cells also proliferated in peripheral blood mononuclear cells (PBMC) stimulated with cytomegalovirus (CMV) Ag. CONCLUSIONS: CD11c expression in CD8(+) T cells reflects anti-tumor CTL activity and would be a marker for immunotherapeutic efficacy in mouse models and probably cancer patients as well.


Subject(s)
CD11c Antigen/metabolism , CD8-Positive T-Lymphocytes/transplantation , Neoplasms, Experimental/therapy , Ovalbumin/administration & dosage , Poly I-C/administration & dosage , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Up-Regulation , Xenograft Model Antitumor Assays
6.
BMC Immunol ; 17(1): 9, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27141827

ABSTRACT

BACKGROUND: Triggering receptors expressed on myeloid cells (Trem) proteins are a family of cell surface receptors used to control innate immune responses such as proinflammatory cytokine production in mice. Trem genes belong to a rapidly expanding family of receptors that include activating and inhibitory paired-isoforms. RESULTS: By comparative genomic analysis, we found that Trem4, Trem5 and Trem-like transcript-6 (Treml6) genes typically paired receptors. These paired Trem genes were murine-specific and originated from an immunoreceptor tyrosine-based inhibition motif (ITIM)-containing gene. Treml6 encoded ITIM, whereas Trem4 and Trem5 lacked the ITIM but possessed positively-charged residues to associate with DNAX activating protein of 12 kDa (DAP12). DAP12 was directly associated with Trem4 and Trem5, and DAP12 coupling was mandatory for their expression on the cell surface. In bone marrow-derived dendritic cells (BMDCs) and macrophages (BMDMs), and splenic DC subsets, polyinosinic-polycytidylic acid (polyI:C) followed by type I interferon (IFN) production induced Trem4 and Treml6 whereas polyI:C or other TLR agonists failed to induce the expression of Trem5. PolyI:C induced Treml6 and Trem4 more efficiently in BMDMs than BMDCs. Treml6 was more potentially up-regulated in conventional DC (cDCs) and plasmacytoid DC (pDCs) than Trem4 in mice upon in vivo stimulation with polyI:C. DISCUSSION: Treml6-dependent inhibitory signal would be dominant in viral infection compared to resting state. Though no direct ligands of these Trem receptors have been determined, the results infer that a set of Trem receptors are up-regulated in response to viral RNA to regulate myeloid cell activation through modulation of DAP12-associated Trem4 and ITIM-containing Treml6.


Subject(s)
Dendritic Cells/immunology , Macrophages/immunology , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cells, Cultured , Female , Gene Expression Regulation , Immunity, Innate , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Domains/genetics , RNA, Double-Stranded/immunology , Receptor, Interferon alpha-beta/genetics , Receptors, Immunologic/genetics
8.
Nat Commun ; 6: 6280, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25692975

ABSTRACT

Ligand stimulation of the Toll-like receptors (TLRs) triggers innate immune response, cytokine production and cellular immune activation in dendritic cells. However, most TLR ligands are microbial constituents, which cause inflammation and toxicity. Toxic response could be reduced for secure immunotherapy through the use of chemically synthesized ligands with defined functions. Here we create an RNA ligand for TLR3 with no ability to activate the RIG-I/MDA5 pathway. This TLR3 ligand is a chimeric molecule consisting of phosphorothioate ODN-guided dsRNA (sODN-dsRNA), which elicits far less cytokine production than poly(I:C) in vitro and in vivo. The activation of TLR3/TICAM-1 pathway by sODN-dsRNA effectively induces natural killer and cytotoxic T cells in tumour-loaded mice, thereby establishing antitumour immunity. Systemic cytokinemia does not occur following subcutaneous or even intraperitoneal administration of sODN-dsRNA, indicating that TICAM-1 signalling with minute local cytokines sufficiently activate dendritic cells to prime tumoricidal effectors in vivo.


Subject(s)
Adjuvants, Immunologic/chemistry , Killer Cells, Natural/cytology , Lymphocyte Activation/drug effects , RNA/chemistry , T-Lymphocytes, Cytotoxic/cytology , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cytokines/metabolism , Dendritic Cells/cytology , Female , HEK293 Cells , HeLa Cells , Humans , Inflammation , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Phosphorothioate Oligonucleotides/chemistry , Poly I-C/chemistry , Signal Transduction
9.
Biochem Biophys Res Commun ; 457(3): 445-50, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25596131

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that exhibit potent immunosuppressive activity. They are increased in tumor-bearing hosts and contribute to tumor development. Toll-like receptors (TLRs) on MDSCs may modulate the tumor-supporting properties of MDSCs through pattern-recognition. Pam2 lipopeptides represented by Pam2CSK4 serve as a TLR2 agonist to exert anti-tumor function by dendritic cell (DC)-priming that leads to NK cell activation and cytotoxic T cell proliferation. On the other hand, TLR2 enhances tumor cell progression/invasion by activating tumor-infiltrating macrophages. How MDSCs respond to TLR2 agonists has not yet been determined. In this study, we found intravenous administration of Pam2CSK4 systemically up-regulated the frequency of MDSCs in EG7 tumor-bearing mice. The frequency of tumor-infiltrating MDSCs was accordingly increased in response to Pam2CSK4. MDSCs were not increased by Pam2CSK4 stimuli in TLR2 knockout (KO) mice. Adoptive transfer experiments using CFSE-labeled MDSCs revealed that the TLR2-positive MDSCs survived long in tumor-bearing mice in response to Pam2CSK4 treatment. Since the increased MDSC population sustained immune-suppressive properties, our study suggests that Pam2CSK4-triggered TLR2 activation enhances the MDSC potential and suppress antitumor immune response in tumor microenvironment.


Subject(s)
Lipopeptides/pharmacology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Toll-Like Receptor 2/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Female , Immunotherapy , Ligands , Lipopeptides/immunology , Lipopeptides/metabolism , Lymphoma/immunology , Lymphoma/metabolism , Lymphoma/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/immunology , Signal Transduction/drug effects , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Tumor Microenvironment
10.
Immunobiology ; 220(1): 74-82, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25257859

ABSTRACT

CD4(+) T cell effectors are crucial for establishing antitumor immunity. Dendritic cell maturation by immune adjuvants appears to facilitate subset-specific CD4(+) T cell proliferation, but the adjuvant effect for CD4 T on induction of cytotoxic T lymphocytes (CTLs) is largely unknown. Self-antigenic determinants with low avidity are usually CD4 epitopes in mutated proteins with tumor-associated class I-antigens (TAAs). In this study, we made a chimeric version of survivin, a target of human CTLs. The chimeric survivin, where human survivin-2B containing a TAA was embedded in the mouse survivin frame (MmSVN2B), was used to immunize HLA-A-2402/K(b)-transgenic (HLA24(b)-Tg) mice. Subcutaneous administration of MmSVN2B or xenogeneic human survivin (control HsSNV2B) to HLA24(b)-Tg mice failed to induce an immune response without co-administration of an RNA adjuvant polyI:C, which was required for effector induction in vivo. Although HLA-A-2402/K(b) presented the survivin-2B peptide in C57BL/6 mice, 2B-specific tetramer assays showed that no CD8(+) T CTLs specific to survivin-2B proliferated above the detection limit in immunized mice, even with polyI:C treatment. However, the CD4(+) T cell response, as monitored by IFN-γ, was significantly increased in mice given polyI:C+MmSVN2B. The Th1 response and antibody production were enhanced in the mice with polyI:C. The CD4 epitope responsible for effector function was not Hs/MmSNV13-27, a nonconserved region between human and mouse survivin, but region 53-67, which was identical between human and mouse survivin. These results suggest that activated, self-reactive CD4(+) helper T cells proliferate in MmSVN2B+polyI:C immunization and contribute to Th1 polarization followed by antibody production, but hardly participate in CTL induction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Inhibitor of Apoptosis Proteins/immunology , Peptide Fragments/immunology , Poly I-C/immunology , Recombinant Fusion Proteins/immunology , Repressor Proteins/immunology , Amino Acid Sequence , Animals , Antibody Formation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Exons , Gene Expression , Gene Order , Genetic Loci , HLA-A24 Antigen/genetics , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Open Reading Frames , Peptide Fragments/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Repressor Proteins/genetics , Survivin
11.
J Innate Immun ; 7(1): 47-58, 2015.
Article in English | MEDLINE | ID: mdl-25115498

ABSTRACT

Toll-like receptors (TLRs) and cytoplasmic RNA sensors have been reported to be involved in the regulation of hepatitis B virus (HBV) replication, but remain controversial due to the lack of a natural infectious model. Our current study sets out to characterize aspects of the role of the innate immune system in eliminating HBV using hydrodynamic-based injection of HBV replicative plasmid and knockout mice deficient in specific pathways of the innate system. The evidence indicated that viral replication was not affected by MAVS or TICAM-1 knockout, but absence of interferon regulatory factor 3 (IRF-3) and IRF-7 transcription factors, as well as the interferon (IFN) receptor, had an adverse effect on the inhibition of HBV replication, demonstrating the dispensability of MAVS and TICAM-1 pathways in the early innate response against HBV. Myd88(-/-) mice did not have a significant increase in the initial viremia, but substantial viral antigen persisted in the mice sera, a response similar to Rag2(-/-) mice, suggesting that the MyD88-dependent pathway participated in evoking an adaptive immune response against the clearance of intrahepatic HBV. Taken together, we show that the RNA-sensing pathways do not participate in the regulation of HBV replication in a mouse model; meanwhile MyD88 is implicated in the HBV clearance.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Vesicular Transport/immunology , Hepatitis B virus/physiology , Hepatitis B/immunology , Signal Transduction/immunology , Virus Replication/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Vesicular Transport/genetics , Animals , Disease Models, Animal , Hepatitis B/genetics , Hepatitis B Antigens/genetics , Hepatitis B Antigens/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics
12.
J Immunol ; 193(10): 5199-207, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25320282

ABSTRACT

Polyinosinic-polycytidylic acid strongly promotes the antitumor activity of NK cells via TLR3/Toll/IL-1R domain-containing adaptor molecule 1 and melanoma differentiation-associated protein-5/mitochondrial antiviral signaling protein pathways. Polyinosinic-polycytidylic acid acts on accessory cells such as dendritic cells (DCs) and macrophages (Mφs) to secondarily activate NK cells. In a previous study in this context, we identified a novel NK-activating molecule, named IFN regulatory factor 3-dependent NK-activating molecule (INAM), a tetraspanin-like membrane glycoprotein (also called Fam26F). In the current study, we generated INAM-deficient mice and investigated the in vivo function of INAM. We found that cytotoxicity against NK cell-sensitive tumor cell lines was barely decreased in Inam(-/-) mice, whereas the number of IFN-γ-producing cells was markedly decreased in the early phase. Notably, deficiency of INAM in NK and accessory cells, such as CD8α(+) conventional DCs and Mφs, led to a robust decrease in IFN-γ production. In conformity with this phenotype, INAM effectively suppressed lung metastasis of B16F10 melanoma cells, which is controlled by NK1.1(+) cells and IFN-γ. These results suggest that INAM plays a critical role in NK-CD8α(+) conventional DC (and Mφ) interaction leading to IFN-γ production from NK cells in vivo. INAM could therefore be a novel target molecule for cancer immunotherapy against IFN-γ-suppressible metastasis.


Subject(s)
Interferon-gamma/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Melanoma, Experimental/genetics , Membrane Glycoproteins/immunology , Poly I-C/pharmacology , Animals , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation , Interferon-gamma/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphocyte Activation , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Transplantation , Signal Transduction
13.
J Immunol ; 192(6): 2770-7, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24532585

ABSTRACT

Hepatitis C virus (HCV) is a major cause of liver disease. The innate immune system is essential for controlling HCV replication, and HCV is recognized by RIG-I and TLR3, which evoke innate immune responses through IPS-1 and TICAM-1 adaptor molecules, respectively. IL-28B is a type III IFN, and genetic polymorphisms upstream of its gene are strongly associated with the efficacy of polyethylene glycol-IFN and ribavirin therapy. As seen with type I IFNs, type III IFNs induce antiviral responses to HCV. Recent studies established the essential role of TLR3-TICAM-1 pathway in type III IFN production in response to HCV infection. Contrary to previous studies, we revealed an essential role of IPS-1 in type III IFN production in response to HCV. First, using IPS-1 knockout mice, we revealed that IPS-1 was essential for type III IFN production by mouse hepatocytes and CD8(+) dendritic cells (DCs) in response to cytoplasmic HCV RNA. Second, we demonstrated that type III IFN induced RIG-I but not TLR3 expression in CD8(+) DCs and augmented type III IFN production in response to cytoplasmic HCV RNA. Moreover, we showed that type III IFN induced cytoplasmic antiviral protein expression in DCs and hepatocytes but failed to promote DC-mediated NK cell activation or cross-priming. Our study indicated that IPS-1-dependent pathway plays a crucial role in type III IFN production by CD8(+) DCs and hepatocytes in response to HCV, leading to cytoplasmic antiviral protein expressions.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cytokines/immunology , Dendritic Cells/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Hepatocytes/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cell Line , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , DEAD-box RNA Helicases/metabolism , Dendritic Cells/metabolism , Dendritic Cells/virology , Flow Cytometry , Gene Expression/drug effects , Gene Expression/immunology , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatocytes/metabolism , Hepatocytes/virology , Host-Pathogen Interactions/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/immunology , Poly I-C/pharmacology , RNA, Viral/genetics , RNA, Viral/immunology , RNA, Viral/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Methods Enzymol ; 535: 149-65, 2014.
Article in English | MEDLINE | ID: mdl-24377923

ABSTRACT

The innate immune system plays key roles in antimicrobial responses by developing the pattern-recognition receptors that recognize microbial components. The endosomal Toll-like receptors (TLRs) and cytosolic RIG-I-like receptors (RLRs) both recognize viral nucleic acids and are essential for antiviral immunity. Recent evidence suggests that compartmentalization of the receptors, and also their adaptor molecule, is important for discrimination between self and nonself and for distinct innate immune signals. TLR3 is a type I transmembrane protein that localizes in the endosomal membrane in myeloid dendritic cells (DCs) and fibroblasts/epithelial cells. TLR3 recognizes extracellular viral double-stranded RNA (dsRNA) and the synthetic dsRNA, poly(I:C). On recognition of dsRNA in the endosomes, TLR3 oligomerizes and induces type I interferon and proinflammatory cytokine production via an adaptor molecule, TICAM-1 (also known as TRIF). Additionally, the TLR3 signal in DCs triggers gene transcription required for DC maturation and the activation of natural killer cells and cytotoxic T lymphocytes. Remarkably, it has been reported that extracellular dsRNA is also recognized by cytosolic RLR. Making a distinction between TLR3-mediated endosomal signaling and RLR-mediated signaling is key to understanding the role of these receptors in innate immunity.


Subject(s)
Endosomes/metabolism , Toll-Like Receptor 3/metabolism , Animals , Cytokines/metabolism , Dendritic Cells/metabolism , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Killer Cells, Natural/physiology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes, Cytotoxic/physiology
15.
Expert Opin Ther Targets ; 17(5): 533-44, 2013 May.
Article in English | MEDLINE | ID: mdl-23414438

ABSTRACT

INTRODUCTION: Many forms of RNA duplexes with agonistic activity for pattern-recognition receptors have been reported, some of which are candidates for adjuvant immunotherapy for cancer. These RNA duplexes induce cytokines, interferons (IFNs) and cellular effectors mainly via two distinct pathways, TLR3/TICAM-1 and MDA5/MAVS. AREAS COVERED: We determined which pathway of innate immunity predominantly participates in evoking tumor immunity in response to RNA adjuvants. EXPERT OPINION: In knockout (KO) mouse studies, robust cytokine or IFN production is dependent on systemic activation of the MAVS pathway, whereas maturation of dendritic cells (DCs) to drive cellular effectors (i.e., NK and CTL) depends on the TICAM-1 pathway in DCs. MAVS activation often causes endotoxin-like cytokinemia, while the TICAM-1 activation does not. Unlike the TLR/MyD88 pathway, this TICAM-1 pathway barely accelerates tumor progression. Although the therapeutic effect in human patients of MAVS-activating or TICAM-1-activating RNA duplexes remains undetermined, the design of a TLR3 agonist with optimized toxicity and dose is an important goal for human immunotherapy. Here we summarize current knowledge on available RNA duplex formulations, and offer a possible approach to developing a promising RNA duplex for clinical tests.


Subject(s)
Neoplasms/immunology , Toll-Like Receptor 3/immunology , Adaptor Proteins, Signal Transducing , Animals , DEAD Box Protein 58 , DEAD-box RNA Helicases , Humans , Immunity, Cellular , Immunotherapy , Interferon-Induced Helicase, IFIH1 , Neoplasms/therapy , Receptors, Immunologic , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism , Tumor Microenvironment
16.
Oncoimmunology ; 1(6): 917-923, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-23162759

ABSTRACT

The engagement of Toll-like receptor 3 (TLR3) leads to the oligomerization of the adaptor TICAM-1 (TRIF), which can induces either of three acute cellular responses, namely, cell survival coupled to Type I interferon production, or cell death, via apoptosis or necrosis. The specific response elicited by TLR3 determines the fate of affected cells, although the switching mechanism between the two cell death pathways in TLR3-stimulated cells remains molecularly unknown. Tumor necrosis factor α (TNFα)-mediated cell death can proceed via apoptosis or via a non-apoptotic pathway, termed necroptosis or programmed necrosis, which have been described in detail. Interestingly, death domain-containing kinases called receptor-interacting protein kinases (RIPs) are involved in the signaling pathways leading to these two cell death pathways. Formation of the RIP1/RIP3 complex (called necrosome) in the absence of caspase 8 activity is crucial for the induction of necroptosis in response to TNFα signaling. On the other hand, RIP1 is known to interact with the C-terminal domain of TICAM-1 and to modulate TLR3 signaling. In macrophages and perhaps tumor cell lines, RIP1/RIP3-mediated necroptotic cell death can ensue the administration of the TLR agonist polyI:C. If this involved the TLR3/TICAM-1 pathway, the innate sensing of viral dsRNA would be linked to cytopathic effects and to persistent inflammation, in turn favoring the release of damage-associated molecular patterns (DAMPs) in the microenvironment. Here, we review accumulating evidence pointing to the involvement of the TLR3/TICAM-1 axis in tumor cell necroptosis and the subsequent release of DAMPs.

17.
Oncoimmunology ; 1(5): 581-592, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22934250

ABSTRACT

PolyI:C is a nucleotide pattern molecule that induces cross-presentation of foreign Ag in myeloid dendritic cells (DC) and MHC Class I-dependent proliferation of cytotoxic T lymphocytes (CTL). DC (BM or spleen CD8α(+)) have sensors for dsRNA including polyI:C to signal facilitating cross-presentation. Endosomal TLR3 and cytoplasmic RIG-I/MDA5 are reportedly responsible for polyI:C sensing and presumed to deliver signal for cross-presentation via TICAM-1 (TRIF) and IPS-1 (MAVS, Cardif, VISA) adaptors, respectively. In fact, when tumor-associated Ag (TAA) was simultaneously taken up with polyI:C in DC, the DC cross-primed CTL specific to the TAA in a syngenic mouse model. Here we tested which of the TICAM-1 or IPS-1 pathway participate in cross-presentation of tumor-associated soluble Ag and retardation of tumor growth in the setting with a syngeneic tumor implant system, EG7/C57BL6, and exogenously challenged soluble Ag (EG7 lysate) and polyI:C. When EG7 lysate and polyI:C were subcutaneously injected in tumor-bearing mice, EG7 tumor growth retardation was observed in wild-type and to a lesser extent IPS-1(-/-) mice, but not TICAM-1(-/-) mice. IRF-3/7 were essential but IPS-1 and type I IFN were minimally involved in the polyI:C-mediated CTL proliferation. Although both TICAM-1 and IPS-1 contributed to CD86/CD40 upregulation in CD8α(+) DC, H2K(b)-SL8 tetramer and OT-1 proliferation assays indicated that OVA-recognizing CD8 T cells predominantly proliferated in vivo through TICAM-1 and CD8α(+) DC is crucial in ex vivo analysis. Ultimately, tumor regresses > 8 d post polyI:C administration. The results infer that soluble tumor Ag induces tumor growth retardation, i.e., therapeutic potential, if the TICAM-1 signal coincidentally occurs in CD8α(+) DC around the tumor.

18.
FASEB J ; 26(10): 4264-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22782974

ABSTRACT

The aging suppressor geneKlotho is predominantly expressed in the kidney irrespective of species. Because Klotho protein is an essential component of an endocrine axis that regulates renal phosphate handling, the kidney-specific expression is biologically relevant; however, little is known about its underlying mechanisms. Here we provide in vitro and in vivo evidence indicating that promoter methylation restricts the expression of the Klotho gene in the kidney. Based on evolutionary conservation and histone methylation patterns, the region up to -1200 bp was defined as a major promoter element of the human Klotho gene. This region displayed promoter activity equally in Klotho-expressing and -nonexpressing cells in transient reporter assays, but the activity was reduced to ∼20% when the constructs were integrated into the chromatin in the latter. Both endogenous and transfected Klotho promoters were 30-40% methylated in Klotho-nonexpressing cells, but unmethylated in Klotho-expressing renal tubular cells. DNA demethylating agents increased Klotho expression 1.5- to 3.0-fold in nonexpressing cells and restored the activity of silenced reporter constructs. Finally, we demonstrated that a severe hypomorphic allele of Klotho had aberrant CpG methylation in kl/kl mice. These findings might be useful in therapeutic intervention for accelerated aging and several complications caused by Klotho down-regulation.


Subject(s)
DNA Methylation/physiology , Glucuronidase/metabolism , Kidney/metabolism , Promoter Regions, Genetic/genetics , Animals , Cell Line , DNA Methylation/genetics , Glucuronidase/genetics , Humans , Immunoblotting , Klotho Proteins , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
19.
J Innate Immun ; 3(3): 264-73, 2011.
Article in English | MEDLINE | ID: mdl-21454965

ABSTRACT

Recent progress in understanding the outcomes of pattern-recognition by myeloid dendritic cells (mDC) allows us to delineate the pathways driving natural killer (NK) cell activation. Mouse mDC mature in response to microbial patterns and are converted to an NK cell-activating phenotype. The MyD88 pathway, the Toll/IL-1 receptor homology domain-containing adaptor molecule (TICAM)-1 (TRIF) pathway, and the interferon (IFN)-ß promoter stimulator 1 (IPS-1) pathway in mDC participate in driving NK activation, as shown by analyses in knockout mice. Studies using synthetic compounds for Toll-like receptors/RIG-I-like receptors have demonstrated that mDC-NK cell contact induces NK cell activation without the participation of cytokines in mice. In vivo bone marrow transplantation analysis revealed that the IPS-1 pathway in nonmyeloid cells and the TICAM-1 pathway in mDC are crucial for dsRNA-mediated in vivo NK activation. These results infer the presence of cytokine-dependent and cytokine-independent modes of NK activation in conjunction with innate immune activation. Here, we focus on the IFN-inducing pathways and mDC-NK contact-induced NK activation and discuss the reported various NK activation modes.


Subject(s)
Dendritic Cells/immunology , Interferon-beta/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Receptors, Pattern Recognition/immunology , Animals , Cell Communication , Humans , Immunity, Innate , Lymphocyte Activation , Mice , Signal Transduction
20.
J Exp Med ; 207(12): 2675-87, 2010 Nov 22.
Article in English | MEDLINE | ID: mdl-21059856

ABSTRACT

In myeloid dendritic cells (mDCs), TLR3 is expressed in the endosomal membrane and interacts with the adaptor toll/interleukin 1 receptor homology domain-containing adaptor molecule 1 (TICAM-1; TRIF). TICAM-1 signals culminate in interferon (IFN) regulatory factor (IRF) 3 activation. Co-culture of mDC pretreated with the TLR3 ligand polyI:C and natural killer (NK) cells resulted in NK cell activation. This activation was triggered by cell-to-cell contact but not cytokines. Using expression profiling and gain/loss-of-function analyses of mDC genes, we tried to identify a TICAM-1-inducing membrane protein that participates in mDC-mediated NK activation. Of the nine candidates screened, one contained a tetraspanin-like sequence and satisfied the screening criteria. The protein, referred to as IRF-3-dependent NK-activating molecule (INAM), functioned in both the mDC and NK cell to facilitate NK activation. In the mDC, TICAM-1, IFN promoter stimulator 1, and IRF-3, but not IRF-7, were required for mDC-mediated NK activation. INAM was minimally expressed on NK cells, was up-regulated in response to polyI:C, and contributed to mDC-NK reciprocal activation via its cytoplasmic tail, which was crucial for the activation signal in NK cells. Adoptive transfer of INAM-expressing mDCs into mice implanted with NK-sensitive tumors caused NK-mediated tumor regression. We identify a new pathway for mDC-NK contact-mediated NK activation that is governed by a TLR signal-derived membrane molecule.


Subject(s)
Dendritic Cells/physiology , Interferon Inducers/pharmacology , Interferon Regulatory Factor-3/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Membrane Glycoproteins/physiology , Poly I-C/pharmacology , Adaptor Proteins, Vesicular Transport/physiology , Amino Acid Sequence , Animals , Cell Line , Interferon-gamma/biosynthesis , Interleukin-15/biosynthesis , Mice , Mice, Inbred C57BL , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL