Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Microbiol Immunol ; 68(2): 65-74, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38105559

ABSTRACT

Cervical cancer is caused mostly by human papillomavirus (HPV), and several HPV vaccines have been developed to prevent its onset. Vaccines include antigens as well as adjuvants, with adjuvants playing an important role in activating the innate immune responses necessary for inducing adaptive immunological responses. Recent research has shown the presence of trained immunity inside the innate immune system. However, trained immunity conferred by HPV vaccinations is not well understood. In this work, we explored the innate immune responses and trained immunity caused by two HPV vaccines, Cervarix and Gardasil. Cervarix includes monophosphoryl lipid A and an aluminum adjuvant, and it significantly increased the expression of IL-6 and IFN-ß mRNAs in RAW264.7 cells. On the contrary, Gardasil, which only includes an aluminum adjuvant, exhibited little cytokine expression but increased the expression of TLRs. Furthermore, Cervarix significantly increased IL-1ß secretion from mouse macrophages, while Gardasil only mildly induced IL-1ß secretion. Interestingly, initial stimulation with Gardasil enhanced the expression of IL-6 and TNF-α mRNAs upon secondary stimulation with TLR ligands, indicating that Gardasil induced trained immunity in macrophages. Moreover, Gardasil injection into mice resulted in enhanced TNF-α production in sera following secondary TLR stimulation. Our findings suggest that HPV vaccinations have the ability to induce trained immunity that modulate TLR ligand responses.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Humans , Animals , Mice , Cytokines , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18 , Tumor Necrosis Factor-alpha , Interleukin-6/genetics , Trained Immunity , Papillomavirus Infections/prevention & control , Aluminum , Papillomavirus Vaccines/genetics , Adjuvants, Immunologic , Toll-Like Receptors
2.
bioRxiv ; 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37397991

ABSTRACT

Post-translational modifications of histone tails alter chromatin accessibility to regulate gene expression. Some viruses exploit the importance of histone modifications by expressing histone mimetic proteins that contain histone-like sequences to sequester complexes that recognize modified histones. Here we identify an evolutionarily conserved and ubiquitously expressed, endogenous mammalian protein Nucleolar protein 16 (NOP16) that functions as a H3K27 mimic. NOP16 binds to EED in the H3K27 trimethylation PRC2 complex and to the H3K27 demethylase JMJD3. NOP16 knockout selectively globally increases H3K27me3, a heterochromatin mark, without altering methylation of H3K4, H3K9, or H3K36 or acetylation of H3K27. NOP16 is overexpressed and linked to poor prognosis in breast cancer. Depletion of NOP16 in breast cancer cell lines causes cell cycle arrest, decreases cell proliferation and selectively decreases expression of E2F target genes and of genes involved in cell cycle, growth and apoptosis. Conversely, ectopic NOP16 expression in triple negative breast cancer cell lines increases cell proliferation, cell migration and invasivity in vitro and tumor growth in vivo , while NOP16 knockout or knockdown has the opposite effect. Thus, NOP16 is a histone mimic that competes with Histone H3 for H3K27 methylation and demethylation. When it is overexpressed in cancer, it derepresses genes that promote cell cycle progression to augment breast cancer growth.

3.
NPJ Vaccines ; 7(1): 16, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35136071

ABSTRACT

mRNA-based vaccines have been used globally to eradicate the coronavirus-disease 2019 (COVID-19) pandemic. Vaccine efficacy and adverse reactions depend on immune responses, such as proinflammatory cytokine production and lymphocyte activation. We conducted a prospective cohort study to investigate relationships among specific antibody titers, adverse reactions, proinflammatory cytokine production, and immune-regulatory microRNA (miRNA) levels in serum extracellular vesicles (EVs) after COVID-19 vaccination (BNT162b2). Local adverse reactions after the second dose, such as local pain and swelling, were less correlated with those of systemic symptoms, such as fever and muscle pain, whereas serum TNF-α levels were associated with systemic adverse reactions and with specific antibody titers. Interestingly, EV miR-92a-2-5p levels in sera were negatively correlated with degrees of adverse reactions, and EV miR-148a levels were associated with specific antibody titers. Our data suggest a potential of circulating EV miRNAs as biomarkers for vaccine efficacy and adverse reactions.

4.
Genome Biol ; 22(1): 134, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33947439

ABSTRACT

BACKGROUND: The evolution of multicellularity is a critical event that remains incompletely understood. We use the social amoeba, Dictyostelium discoideum, one of the rare organisms that readily transits back and forth between both unicellular and multicellular stages, to examine the role of epigenetics in regulating multicellularity. RESULTS: While transitioning to multicellular states, patterns of H3K4 methylation and H3K27 acetylation significantly change. By combining transcriptomics, epigenomics, chromatin accessibility, and orthologous gene analyses with other unicellular and multicellular organisms, we identify 52 conserved genes, which are specifically accessible and expressed during multicellular states. We validated that four of these genes, including the H3K27 deacetylase hdaD, are necessary and that an SMC-like gene, smcl1, is sufficient for multicellularity in Dictyostelium. CONCLUSIONS: These results highlight the importance of epigenetics in reorganizing chromatin architecture to facilitate multicellularity in Dictyostelium discoideum and raise exciting possibilities about the role of epigenetics in the evolution of multicellularity more broadly.


Subject(s)
Dictyostelium/cytology , Dictyostelium/genetics , Epigenesis, Genetic , Acetylation , Animals , Caenorhabditis elegans/cytology , Chromatin/metabolism , Gene Expression Profiling , Histones/metabolism , Methylation , Schizosaccharomyces/cytology , Transcription Factors/metabolism
5.
Uirusu ; 71(1): 33-40, 2021.
Article in Japanese | MEDLINE | ID: mdl-35526992
6.
Mol Cell ; 75(3): 631-643.e8, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31279658

ABSTRACT

mRNAs are regulated by nucleotide modifications that influence their cellular fate. Two of the most abundant modified nucleotides are N6-methyladenosine (m6A), found within mRNAs, and N6,2'-O-dimethyladenosine (m6Am), which is found at the first transcribed nucleotide. Distinguishing these modifications in mapping studies has been difficult. Here, we identify and biochemically characterize PCIF1, the methyltransferase that generates m6Am. We find that PCIF1 binds and is dependent on the m7G cap. By depleting PCIF1, we generated transcriptome-wide maps that distinguish m6Am and m6A. We find that m6A and m6Am misannotations arise from mRNA isoforms with alternative transcription start sites (TSSs). These isoforms contain m6Am that maps to "internal" sites, increasing the likelihood of misannotation. We find that depleting PCIF1 does not substantially affect mRNA translation but is associated with reduced stability of a subset of m6Am-annotated mRNAs. The discovery of PCIF1 and our accurate mapping technique will facilitate future studies to characterize m6Am's function.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Nuclear Proteins/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , Transcriptome/genetics , Adenosine/genetics , Humans , Methylation , Methyltransferases/genetics , Protein Biosynthesis/genetics , Transcription Initiation Site
7.
Cancer Sci ; 109(7): 2119-2129, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29791768

ABSTRACT

Immunological checkpoint blockade therapies benefit a limited population of cancer patients. We have previously shown that vaccine immunotherapy with Toll-like receptor (TLR)3-adjuvant and tumor antigen overcomes anti-programmed death ligand-1 (PD-L1) resistance in mouse tumor models. In the present study, 4 different ovalbumin (OVA)-expressing tumor cell lines were implanted into syngeneic mice and subjected to anti-tumor immunotherapy using ARNAX and whole OVA protein. ARNAX is a TLR3-specific agonist that does not activate the mitochondrial antiviral-signaling protein (MAVS) pathway, and thus does not induce systemic inflammation. Dendritic cell priming and proliferative CTL were induced by ARNAX + OVA, but complete remission was achieved only in a PD-L1-low cell line of EG7. Addition of anti-PD-L1 antibody to the ARNAX + OVA therapy brought complete remission to another PD-L1-high subline of EG7. Tumor shrinkage but not remission was observed in MO5 in that regimen. We analyzed tumor cells and tumor-infiltrating immune cells to identify factors associated with successful ARNAX vaccine therapy. Tumors that responded to ARNAX therapy expressed high levels of MHC class I and low levels of PD-L1. The tumor-infiltrating immune cells in ARNAX-susceptible tumors contained fewer immunosuppressive myeloid cells with low PD-L1 expression. Combination with anti-PD-L1 antibody functioned not only within tumor sites but also within lymphoid tissues, augmenting the therapeutic efficacy of the ARNAX vaccine. Notably, ARNAX therapy induced memory CD8+ T cells and rejection of reimplanted tumors. Thus, ARNAX vaccine + anti-PD-L1 therapy enabled permanent remission against some tumors that stably present antigens.


Subject(s)
Immunotherapy/methods , T-Lymphocytes/immunology , Toll-Like Receptor 3/agonists , Animals , Antigens, Neoplasm/immunology , Disease Models, Animal , Immunologic Memory/immunology , Mice , Mice, Inbred C57BL , Tumor Microenvironment/immunology
8.
Biochem Biophys Res Commun ; 499(4): 985-991, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29627569

ABSTRACT

Stimulator of interferon genes (STING) is an essential molecule for the production of type I interferon (IFN), and other inflammatory cytokines, in response to cytosolic DNA. STING contributes to host defense against infection and anti-tumor responses. Previous reports have demonstrated that STING signaling is required by the adaptor Toll-IL-1 receptor-containing adaptor molecule-1 (TICAM-1), which has been identified as a TLR3-adaptor molecule using mouse embryonic fibroblasts. Here, we demonstrate that TICAM-1 does not affect STING-mediated innate immune responses, as increases in the mRNA expression levels of IFN-ß, IL-6, and CCL5 were observed in bone marrow-derived or splenic myeloid cells. Moreover, STING ligand-enhanced co-stimulatory molecule expression, including CD80, CD86, and CD40, was detected on splenic CD11c + DCs, even in Ticam-1-deficient mice. Our results suggest that STING-mediated innate immune responses and dendritic cell maturation do not require TICAM-1 in myeloid lineage immune cells. TICAM-1 is ubiquitously expressed, even in cell types which do not express TLR3. Therefore, TICAM-1 may possess different functions depending on cell type and signaling purposes.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Immunity, Innate , Membrane Proteins/metabolism , Myeloid Cells/immunology , Animals , Bone Marrow/metabolism , Cytokines/metabolism , Dendritic Cells/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Inflammation Mediators/metabolism , Ligands , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Spleen/cytology , Toll-Like Receptor 3/metabolism
9.
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
10.
Cancer Sci ; 109(4): 956-965, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29465830

ABSTRACT

Radiotherapy induces anti-tumor immunity by induction of tumor antigens and damage-associated molecular patterns (DAMP). DNA, a representative DAMP in radiotherapy, activates the stimulator of interferon genes (STING) pathway which enhances the immune response. However, the immune response does not always parallel the inflammation associated with radiotherapy. This lack of correspondence may, in part, explain the radiation-resistance of tumors. Additive immunotherapy is expected to revive tumor-specific CTL facilitating radiation-resistant tumor shrinkage. Herein pre-administration of the double-stranded RNA, polyinosinic-polycytidylic acid (polyI:C), in conjunction with radiotherapy, was shown to foster tumor suppression in mice bearing radioresistant, ovalbumin-expressing Lewis lung carcinoma (LLC). Extrinsic injection of tumor antigen was not required for tumor suppression. No STING- and CTL-response was induced by radiation in the implant tumor. PolyI:C was more effective for induction of tumor growth retardation at 1 day before radiation than at post-treatment. PolyI:C targeted Toll-like receptor 3 with minimal effect on the mitochondrial antiviral-signaling protein pathway. Likewise, the STING pathway barely contributed to LLC tumor suppression. PolyI:C primed antigen-presenting dendritic cells in draining lymph nodes to induce proliferation of antigen-specific CTL. By combination therapy, CTL efficiently infiltrated into tumors with upregulation of relevant chemokine transcripts. Batf3-positive DC and CD8+ T cells were essential for therapeutic efficacy. Furthermore, polyI:C was shown to stimulate tumor-associated macrophages and release tumor necrosis factor alpha, which acted on tumor cells and increased sensitivity to radiation. Hence, polyI:C treatment prior to radiotherapy potentially induces tumor suppression by boosting CTL-dependent and macrophage-mediated anti-tumor responses. Eventually, polyI:C and radiotherapy in combination would be a promising therapeutic strategy for radiation-resistant tumors.


Subject(s)
Carcinoma, Lewis Lung/radiotherapy , Cell Proliferation/radiation effects , Toll-Like Receptor 3/metabolism , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy/methods , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/radiation effects , Disease Models, Animal , Immunotherapy, Adoptive/methods , Macrophages/drug effects , Macrophages/metabolism , Macrophages/radiation effects , Mice , Mice, Inbred C57BL , Poly I-C/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/radiation effects
11.
J Innate Immun ; 10(1): 44-55, 2018.
Article in English | MEDLINE | ID: mdl-29069650

ABSTRACT

Melanoma differentiation-associated gene 5 (MDA5) is a pattern recognition receptor that recognizes cytoplasmic viral double-stranded RNA (dsRNA) and initiates rapid innate antiviral responses. MDA5 forms a filament-like multimer along the dsRNA leading to oligomerization, which in turn activates the adaptor protein mitochondrial antiviral signaling protein (MAVS) to provide a signal platform for the induction of type I interferon (IFN) and proinflammatory cytokines. The conformational switch of MDA5 causes antiviral defense, but excessive activation of the MDA5-MAVS pathway may result in autoimmune diseases. The regulatory mechanisms of MDA5 activation remain largely unknown. By yeast 2-hybrid, we identified DNAJB1, a member of the HSP40 (heat shock protein 40) family, as an MDA5-binding protein. HSP40s usually cowork with HSP70s. We found that dsRNA stimulation with physiological conditions upregulated the expression levels of DNAJB1 and HSP70; then the proteins were coupled and translocated into the stress granules, where MDA5 encounters dsRNA. DNAJB1 disrupted MDA5 multimer formation, resulting in the suppression of type I IFN induction. The disruption of endogenous DNAJB1 increased MDA5- and MAVS-mediated IFN promoter activation and rendered cells virus resistant. HSP70 inhibitor also enhanced the IFN-inducing function of MDA5 and MAVS. These results suggest that the DNAJB1-HSP70 complex functions for the natural maintenance of RNA sensing by interacting with MDA5/MAVS.


Subject(s)
HSP40 Heat-Shock Proteins/metabolism , Inflammation/immunology , Interferon-Induced Helicase, IFIH1/metabolism , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Virus Diseases/immunology , Adaptor Proteins, Signal Transducing/metabolism , Autoimmunity , Gene Expression Regulation , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Protein Binding , Protein Multimerization , Signal Transduction
12.
Med Sci (Basel) ; 5(4)2017 Dec 18.
Article in English | MEDLINE | ID: mdl-29258267

ABSTRACT

Induction of immunoglobulin (Ig) A in the mucosa of the upper respiratory tract and the nasal cavity protects against influenza virus infection. Cyclic dinucleotides (CDNs) are used as mucosal adjuvants to enhance the immunogenicity of intranasal influenza hemagglutinin (HA) vaccines. The adjuvant activity of 2'3' cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) on Ig production was investigated in nasal-associated lymphoid tissue (NALT), serum of wild-type C57BL/6J, and stimulator of interferon genes (STING)-deficient mice, which do not recognize cGAMP. Mice were vaccinated intranasally with a HA vaccine with or without the cGAMP adjuvant. IgA and IgG production, T-cell responses, germinal center formation, and cytokine expression in NALT were assayed. cGAMP enhanced IgA and IgG production, and promoted T-cell responses. Intranasal administration of cGAMP activated both NALT and systemic immune cells, induced a favorable cytokine environment for IgA induction, and promoted germinal center formation. The cGAMP effect was STING-dependent. Taken together, cGAMP as an HA vaccine adjuvant promoted a STING-dependent NALT environment suitable for the enhancement of IgA production.

13.
J Biomed Sci ; 24(1): 79, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29041928

ABSTRACT

BACKGROUND: Intestinal tumorigenesis is promoted by myeloid differentiation primary response gene 88 (MyD88) activation in response to the components of microbiota in Apc Min/+ mice. Microbiota also contains double-stranded RNA (dsRNA), a ligand for TLR3, which activates the toll-like receptor adaptor molecule 1 (TICAM-1, also known as TRIF) pathway. METHODS: We established Apc Min/+ Ticam1 -/- mice and their survival was compared to survival of Apc Min/+ Myd88 -/- and wild-type (WT) mice. The properties of polyps were investigated using immunofluorescence staining and RT-PCR analysis. RESULTS: We demonstrate that TICAM-1 is essential for suppression of polyp formation in Apc Min/+ mice. TICAM-1 knockout resulted in shorter survival of mice compared to WT mice or mice with knockout of MyD88 in the Apc Min/+ background. Polyps were more frequently formed in the distal intestine of Apc Min/+ Ticam1 -/- mice than in Apc Min/+ mice. Infiltration of immune cells such as CD11b+ and CD8α+ cells into the polyps was detected histologically. CD11b and CD8α mRNAs were increased in polyps of Apc Min/+ Ticam1 -/- mice compared to Apc Min/+ mice. Gene expression of inducible nitric oxide synthase (iNOS), interferon (IFN)-γ, CXCL9 and IL-12p40 was increased in polyps of Apc Min/+ Ticam1 -/- mice. mRNA and protein expression of c-Myc, a critical transcription factor for inflammation-associated polyposis, were increased in polyps of Apc Min/+ Ticam1 -/- mice. A Lactobacillus strain producing dsRNA was detected in feces of Apc Min/+ mice. CONCLUSION: These results imply that the TLR3/TICAM-1 pathway inhibits polyposis through suppression of c-Myc expression and supports long survival in Apc Min/+ mice.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Colonic Polyps/genetics , Colorectal Neoplasms/genetics , Intestinal Polyps/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Toll-Like Receptor 3/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 3/metabolism
14.
Biochem Biophys Res Commun ; 478(4): 1764-71, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27608599

ABSTRACT

An interferon-inducing DNA sensor STING participates in tumor rejection in mouse models. Here we examined what mechanisms contribute to STING-dependent growth retardation of B16 melanoma sublines by NK cells in vivo. The studies were designed using WT and STING KO black mice, and B16D8 (an NK-sensitive melanoma line having STING) and STING KO B16D8 sublines established for this study. The results from tumor-implant studies suggested that STING in host immune cells and tumor cells induced distinct profiles of chemokines including CXCL10, CCL5 and IL-33, and both participated in NK cell infiltration and activation in B16D8 tumor. Spontaneous activation of STING occurs in host-immune and tumor cells of this NK-sensitive tumor, thereby B16D8 tumor growth being suppressed in this model. Our data show that STING induces tumor cytotoxicity by NK cells through tumor and host immune cell network to contribute to innate surveillance and suppression of tumors in vivo.


Subject(s)
Cell Proliferation/genetics , Killer Cells, Natural/metabolism , Melanoma, Experimental/genetics , Membrane Proteins/genetics , Tumor Burden/genetics , Animals , Blotting, Western , Cell Line, Tumor , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Female , Gene Expression Regulation, Neoplastic , Interferon-beta/genetics , Interferon-beta/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Cancer Sci ; 106(12): 1659-68, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26395101

ABSTRACT

Immune-enhancing adjuvants usually targets antigen (Ag)-presenting cells to tune up cellular and humoral immunity. CD141(+) dendritic cells (DC) represent the professional Ag-presenting cells in humans. In response to microbial pattern molecules, these DCs upgrade the maturation stage sufficient to improve cross-presentation of exogenous Ag, and upregulation of MHC and costimulators, allowing CD4/CD8 T cells to proliferate and liberating cytokines/chemokines that support lymphocyte attraction and survival. These DCs also facilitate natural killer-mediated cell damage. Toll-like receptors (TLRs) and their signaling pathways in DCs play a pivotal role in DC maturation. Therefore, providing adjuvants in addition to Ag is indispensable for successful vaccine immunotherapy for cancer, which has been approved in comparison with antimicrobial vaccines. Mouse CD8α(+) DCs express TLR7 and TLR9 in addition to the TLR2 family (TLR1, 2, and 6) and TLR3, whereas human CD141(+) DCs exclusively express the TLR2 family and TLR3. Although human and mouse plasmacytoid DCs commonly express TLR7/9 to respond to their agonists, the results on mouse adjuvant studies using TLR7/9 agonists cannot be simply extrapolated to human adjuvant immunotherapy. In contrast, TLR2 and TLR3 are similarly expressed in both human and mouse Ag-presenting DCs. Bacillus Calmette-Guerin peptidoglycan and polyinosinic-polycytidylic acid are representative agonists for TLR2 and TLR3, respectively, although they additionally stimulate cytoplasmic sensors: their functional specificities may not be limited to the relevant TLRs. These adjuvants have been posted up to a certain achievement in immunotherapy in some cancers. We herein summarize the history and perspectives of TLR2 and TLR3 agonists in vaccine-adjuvant immunotherapy for cancer.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/immunology , Immunotherapy/methods , Toll-Like Receptor 2/agonists , Toll-Like Receptor 3/agonists , Animals , Humans
17.
Cell Rep ; 11(2): 192-200, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25865883

ABSTRACT

MDA5 is a cytoplasmic viral double-stranded RNA (dsRNA) sensor and triggers type I interferon (IFN) production. MDA5 assembles along viral dsRNA, leading to the formation of an MDA5 filament required for activating the MAVS adaptor. A recent study has revealed that PP1α and PP1γ phosphatases are responsible for dephosphorylating MDA5 and are essential for its activation. Here, we identified RIO kinase 3 (RIOK3) as a protein kinase that phosphorylates the MDA5 C-terminal region. RIOK3 knockout strongly enhanced type I IFN and IFN-inducible gene expression following measles virus infection. Conversely, the ectopic expression of RIOK3 or a phosphomimetic MDA5-S828D mutation attenuated MDA5-mediated signaling. Moreover, RIOK3-mediated MDA5 phosphorylation impaired MDA5 multimer formation, indicating that MDA5 C-terminal phosphorylation interferes with MDA5 filament formation and suppresses its signaling. Our data revealed a regulatory mechanism underlying the activation of the cytoplasmic viral RNA sensor MDA5 in both uninfected and virus-infected cells.


Subject(s)
DEAD-box RNA Helicases/genetics , Immunity, Innate/genetics , Measles/genetics , Protein Serine-Threonine Kinases/genetics , DEAD-box RNA Helicases/biosynthesis , HEK293 Cells , Humans , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon-Induced Helicase, IFIH1 , Measles/virology , Measles virus/genetics , Measles virus/pathogenicity , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , RNA, Double-Stranded/genetics , RNA, Viral/genetics
18.
Anat Sci Int ; 78(4): 228-42, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14686478

ABSTRACT

We investigated the topographical anatomy of the pelvic fasciae and autonomic nerves using macroscopic slices of five decalcified female pelves. The lateral aspect of the supravaginal cervix uteri and superior-most vagina issued abundant thick fiber bundles. These visceral fibrous tissues extended dorsolaterally, joined another fibrous tissue from the rectum (the actual lateral ligament of the rectum) and attached to the parietal fibrous tissues at and around the sciatic foramina (i.e. the sacrospinous ligament, thick fasciae of the coccygeus and piriformis and dorsal end of the covering fascia of the levator ani). The inferior or ventral vagina also issued thick fiber bundles communicating with the levator ani fascia. This connection between the vagina and levator fascia, when stretched, seemed to provide a macroscopic morphology called the arcus tendineus fasciae pelvis. The overall morphology of the visceroparietal fascial bridge exhibited a bilateral wing-like shape. The fascial bridge complex was adjacent but dorso-inferior to the internal iliac vascular sheath and located slightly ventral to the pelvic splanchnic nerve. However, the pelvic plexus and its peripheral branches were embedded in the fascial complex. The hypogastric nerve ran along and beneath the uterosacral peritoneal fold, which did not contain thick fibrous tissue. During surgery, in combination with the superficially located vascular sheath, the morphology of the visceroparietal fascial bridge and associated nerves seemed to be artificially changed and developed into the so-called cardinal, uterosacral, uterovesical and/or rectal lateral ligaments. The classical and original concepts of these pelvic fascial structures may need to be altered to adjust to these surgical observations.


Subject(s)
Autonomic Nervous System/anatomy & histology , Fascia/anatomy & histology , Genitalia, Female/anatomy & histology , Pelvis/anatomy & histology , Aged , Aged, 80 and over , Cadaver , Female , Humans , Magnetic Resonance Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...