Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 534(7607): 402-6, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27281199

RESUMEN

Successful treatment of many patients with advanced cancer using antibodies against programmed cell death 1 (PD-1; also known as PDCD1) and its ligand (PD-L1; also known as CD274) has highlighted the critical importance of PD-1/PD-L1-mediated immune escape in cancer development. However, the genetic basis for the immune escape has not been fully elucidated, with the exception of elevated PD-L1 expression by gene amplification and utilization of an ectopic promoter by translocation, as reported in Hodgkin and other B-cell lymphomas, as well as stomach adenocarcinoma. Here we show a unique genetic mechanism of immune escape caused by structural variations (SVs) commonly disrupting the 3' region of the PD-L1 gene. Widely affecting multiple common human cancer types, including adult T-cell leukaemia/lymphoma (27%), diffuse large B-cell lymphoma (8%), and stomach adenocarcinoma (2%), these SVs invariably lead to a marked elevation of aberrant PD-L1 transcripts that are stabilized by truncation of the 3'-untranslated region (UTR). Disruption of the Pd-l1 3'-UTR in mice enables immune evasion of EG7-OVA tumour cells with elevated Pd-l1 expression in vivo, which is effectively inhibited by Pd-1/Pd-l1 blockade, supporting the role of relevant SVs in clonal selection through immune evasion. Our findings not only unmask a novel regulatory mechanism of PD-L1 expression, but also suggest that PD-L1 3'-UTR disruption could serve as a genetic marker to identify cancers that actively evade anti-tumour immunity through PD-L1 overexpression.


Asunto(s)
Regiones no Traducidas 3'/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Receptor de Muerte Celular Programada 1/genética , Escape del Tumor/genética , Regulación hacia Arriba , Adenocarcinoma/genética , Animales , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Sistemas CRISPR-Cas , Línea Celular Tumoral , Selección Clonal Mediada por Antígenos , Femenino , Marcadores Genéticos/genética , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Linfoma de Células B Grandes Difuso/genética , Ratones , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/biosíntesis , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Neoplasias Gástricas/genética
2.
J Biol Chem ; 294(50): 18969-18979, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31615841

RESUMEN

Cytoplasmic dsRNA is recognized by RNA helicase RIG-I (RIG-I) and melanoma differentiation-associated protein 5 (MDA5), triggering induction of the innate immune response via the mitochondrial antiviral signaling protein (MAVS). In contrast, extracellular dsRNA is internalized into endosomes and recognized by Toll-like receptor 3 (TLR3), which triggers signaling via the Toll-like receptor adaptor molecule 1 (TICAM-1). Poly(I:C) is a synthetic dsRNA analog and increases the expression of octamer-binding protein 3/4 (OCT3/4), NANOG, and SRY-box (SOX) mRNAs during pluripotency induction. However, the mechanism underlying this increase is unclear. Here, we focused on the mechanism of poly(I:C)-induced expression of stem cell-specific genes in human somatic cells. Addition of poly(I:C) to human fibroblast culture medium did not increase OCT3/4 mRNA expression, but poly(I:C) transfection markedly increased OCT3/4 expression and induced nuclear localization of the OCT3/4 protein, implying that not TLR3, but RIG-I and MDA5 are required for OCT3/4 expression. Moreover, although cytoplasmic dsRNA increased OCT3/4 mRNA, cytoplasmic dsDNAs, such as salmon sperm DNA and poly(dA:dT), did not. Interestingly, the expression of NANOG, SOX2, Krüppel-like factor 4 (KLF4), and proto-oncogene c-Myc was also increased by cytoplasmic dsRNA. Of note, siRNAs that silenced MAVS and interferon regulatory factor 1 (IRF1) expression reduced OCT3/4 levels after stimulation with poly(I:C); however, an NF-κB inhibitor and siRNA-mediated knockdown of proto-oncogene c-Jun did not significantly reduce the mRNA levels. We conclude that cytoplasmic dsRNA increases the expression of stem cell-specific genes in human somatic cells in a MAVS- and IRF1-dependent manner.


Asunto(s)
Citoplasma/química , Proteína Homeótica Nanog/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteínas de Transporte de Catión Orgánico/genética , ARN Bicatenario/metabolismo , ARN Mensajero/genética , Diferenciación Celular , Línea Celular , Citoplasma/metabolismo , Humanos , Factor 4 Similar a Kruppel , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Proto-Oncogenes Mas , ARN Mensajero/metabolismo
3.
PLoS Pathog ; 13(1): e1006162, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28103322

RESUMEN

The Tax protein of human T-cell leukemia virus type 1 (HTLV-1) is crucial for the development of adult T-cell leukemia (ATL), a highly malignant CD4+ T cell neoplasm. Among the multiple aberrant Tax-induced effects on cellular processes, persistent activation of transcription factor NF-κB, which is activated only transiently upon physiological stimulation, is essential for leukemogenesis. We and others have shown that Tax induces activation of the IκB kinase (IKK) complex, which is a critical step in NF-κB activation, by generating Lys63-linked polyubiquitin chains. However, the molecular mechanism underlying Tax-induced IKK activation is controversial and not fully understood. Here, we demonstrate that Tax recruits linear (Met1-linked) ubiquitin chain assembly complex (LUBAC) to the IKK complex and that Tax fails to induce IKK activation in cells that lack LUBAC activity. Mass spectrometric analyses revealed that both Lys63-linked and Met1-linked polyubiquitin chains are associated with the IKK complex. Furthermore, treatment of the IKK-associated polyubiquitin chains with Met1-linked-chain-specific deubiquitinase (OTULIN) resulted in the reduction of high molecular weight polyubiquitin chains and the generation of short Lys63-linked ubiquitin chains, indicating that Tax can induce the generation of Lys63- and Met1-linked hybrid polyubiquitin chains. We also demonstrate that Tax induces formation of the active macromolecular IKK complex and that the blocking of Tax-induced polyubiquitin chain synthesis inhibited formation of the macromolecular complex. Taken together, these results lead us to propose a novel model in which the hybrid-chain-dependent oligomerization of the IKK complex triggered by Tax leads to trans-autophosphorylation-mediated IKK activation.


Asunto(s)
Activación Enzimática/fisiología , Productos del Gen tax/metabolismo , Infecciones por HTLV-I/metabolismo , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Immunoblotting , Inmunoprecipitación , Células Jurkat , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Transfección
4.
Cancer Sci ; 109(7): 2119-2129, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29791768

RESUMEN

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.


Asunto(s)
Inmunoterapia/métodos , Linfocitos T/inmunología , Receptor Toll-Like 3/agonistas , Animales , Antígenos de Neoplasias/inmunología , Modelos Animales de Enfermedad , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral/inmunología
5.
Cancer Sci ; 109(4): 956-965, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29465830

RESUMEN

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.


Asunto(s)
Carcinoma Pulmonar de Lewis/radioterapia , Proliferación Celular/efectos de la radiación , Receptor Toll-Like 3/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/efectos de la radiación , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada/métodos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efectos de la radiación , Modelos Animales de Enfermedad , Inmunoterapia Adoptiva/métodos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Poli I-C/farmacología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/efectos de la radiación
6.
Biochem Biophys Res Commun ; 499(4): 985-991, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29627569

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Células Mieloides/inmunología , Animales , Médula Ósea/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Mediadores de Inflamación/metabolismo , Ligandos , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Bazo/citología , Receptor Toll-Like 3/metabolismo
7.
Biochem Biophys Res Commun ; 506(4): 1019-1025, 2018 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-30404733

RESUMEN

Adjuvant stimulates pattern-recognition receptors (PRRs) expressed by dendritic cells, which causes immune-enhancing of T lymphocytes. Adjuvant also induces innate immune response in whole-body cells via PRRs to evoke cytokinemia. A cytokine-mediated immune response is important for the systemic protection of a host from microbial infections. Using an influenza subcomponent vaccine in a mouse model, we intranasally administered a TLR3-specific adjuvant ARNAX + HA split vaccine to mice. ARNAX efficiently induced mucosal IgA and systemic IgG production by nasal drop. Moreover, ARNAX + HA simultaneously induced CD8 and CD4 T cell activation. We have previously shown that ARNAX does not induce harmful systemic cytokine production. Thus, our findings indicate that the ARNAX + HA vaccine is a harmless prophylactic vaccine for flu that induces HA-specific T cell activation and IgA/IgG production. These results suggested that ARNAX + antigen enhanced the immune response without inducing inflammatory toxicity for vaccination against infectious diseases.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Inmunidad Mucosa/efectos de los fármacos , Inmunoglobulina A/biosíntesis , Inmunoglobulina G/biosíntesis , Vacunas contra la Influenza/inmunología , Vacunación , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Tejido Linfoide/patología , Ratones Endogámicos C57BL , Nariz/patología , Poli I-C/farmacología , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Receptor Toll-Like 3/metabolismo
8.
J Immunol ; 196(9): 3865-76, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27022195

RESUMEN

The clathrin-dependent endocytic pathway is crucial for endosomal TLR3- and TLR4-mediated Toll-IL-1R domain-containing adaptor molecule-1 (TICAM-1) signaling. TLR4 uses a different signaling platform, plasma membrane and endosomes, for activation of TIRAP-MyD88 and TICAM-2-TICAM-1, respectively. LPS-induced endocytosis of TLR4 is mandatory for TICAM-1-mediated signaling including IFN-ß production. Several molecules/mechanisms such as CD14, clathrin, and phosphatidylinositol metabolism have been reported to act as inducers of TLR4 translocation. However, the molecular mechanism of spatiotemporal regulation of TLR4 signaling remains unresolved. We have previously shown that Raftlin is essential for clathrin-dependent endocytosis of TLR3 ligand in human epithelial cells and myeloid dendritic cells (DCs). In this article, we demonstrate that Raftlin also mediated LPS-induced TLR4 internalization and TICAM-1 signaling in human monocyte-derived DCs and macrophages (Mo-Mϕs). When Raftlin was knocked down, LPS-induced TLR4-mediated IFN-ß promoter activation, but not NF-κB activation, was decreased in HEK293 cells overexpressing TLR4/MD-2 or TLR4/MD-2/CD14. LPS-induced IFN-ß production by monocyte-derived DCs and Mo-Mϕs was significantly decreased by knockdown of Raftlin. Upon LPS stimulation, Raftlin moved from the cytoplasm to the plasma membrane in Mo-Mϕs, where it colocalized with TLR4. Raftlin associated with clathrin-associated adaptor protein-2 in resting cells and transiently bound to TLR4 and clathrin at the cell surface in response to LPS. Thus, Raftlin appears to modulate cargo selection as an accessary protein of clathrin-associated adaptor protein-2 in clathrin-mediated endocytosis of TLR3/4 ligands.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Células Epiteliales/fisiología , Proteínas de la Membrana/metabolismo , Células Mieloides/fisiología , Animales , Clatrina/metabolismo , Endocitosis , Células HEK293 , Humanos , Interferón beta/metabolismo , Lipopolisacáridos/inmunología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Transducción de Señal , Receptor Toll-Like 4/metabolismo
9.
Proc Jpn Acad Ser B Phys Biol Sci ; 94(3): 153-160, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29526974

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Células Dendríticas/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Animales , Humanos
10.
Biochem Soc Trans ; 45(4): 929-935, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28630139

RESUMEN

Toll-like receptor 4 (TLR4) recognizes lipopolysaccharide (LPS), produces pro-inflammatory cytokines and type I interferons, and associates with a trigger of endotoxin shock. TLR4 is interacted with a TIR domain-containing adaptor molecule-2 (TICAM-2)/TRAM [TRIF (TIR domain-containing adaptor-inducing interferon-ß)-related adaptor molecule] via its Toll-interleukin-1 receptor homology (TIR) domain. TICAM-2 acts as a scaffold protein and activates TIR domain-containing adaptor molecule-1 (TICAM-1)/TRIF. According to the structural analysis by NMR, TICAM-2 interacts with TICAM-1 by the acidic amino acids motif, E87/D88/D89. The TIR domain of TICAM-2 couples with the dimer of TIR domain of TLR4 beneath the membrane, and TICAM-2 itself also forms dimer and constitutes a binding site with TICAM-1. Endosomal localization of TICAM-2 is essential for TLR4-mediated type I interferon-inducing signal from the endosome. N-terminal myristoylation allows TICAM-2 to anchor to the endosomal membrane. Additionally, we have identified two acidic amino acids, D91/E92, as a functional motif that cooperatively determines endosomal localization of TICAM-2. This structural information of TICAM-2 suggests that the specific structure is indispensable for the endosomal localization and type I interferon production of TICAM-2. Taken together with the knowledge on cytoplasmic sensors for LPS, TICAM-2/TICAM-1 may conform to a signal network on TLR4 to facilitate induction of cytokine disorders.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Endosomas/metabolismo , Modelos Biológicos , Procesamiento Proteico-Postraduccional , Transducción de Señal , Receptor Toll-Like 4/agonistas , Acilación , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras del Transporte Vesicular/química , Secuencias de Aminoácidos , Animales , Dimerización , Humanos , Interferón Tipo I/metabolismo , Ácido Mirístico/metabolismo , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Transporte de Proteínas , Receptor Toll-Like 4/química , Receptor Toll-Like 4/metabolismo
11.
J Biomed Sci ; 24(1): 79, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29041928

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Pólipos del Colon/genética , Neoplasias Colorrectales/genética , Pólipos Intestinales/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Receptor Toll-Like 3/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 3/metabolismo
12.
Microbiol Immunol ; 61(3-4): 107-113, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28370181

RESUMEN

Viruses usually exhibit strict species-specificity as a result of co-evolution with the host. Thus, in mouse models, a great barrier exists for analysis of infections with human-tropic viruses. Mouse models are unlikely to faithfully reproduce the human immune response to viruses or viral compounds and it is difficult to evaluate human therapeutic efficacy with antiviral reagents in mouse models. Humans and mice essentially have different immune systems, which makes it difficult to extrapolate mouse results to humans. In addition, apart from immunological reasons, viruses causing human diseases do not always infect mice because of species tropism. One way to determine tropism would be a virus receptor that is expressed on affected cells. The development of gene-disrupted mice and Tg mice, which express human receptor genes, enables us to analyze several viral infections in mice. Mice are, indeed, susceptible to human viruses when artificially infected in receptor-supplemented mice. Although the mouse cells less efficiently permit viral replication than do human cells, the models for analysis of human viruses have been established in vivo as well as in vitro, and explain viral pathogenesis in the mouse systems. In most systems, however, nucleic acid sensors and type I interferon suppress viral propagation to block the appearance of infectious manifestation. We herein review recent insight into in vivo antiviral responses induced in mouse infection models for typical human viruses.


Asunto(s)
Modelos Animales de Enfermedad , Ratones , Virosis/patología , Virosis/virología , Animales , Animales Modificados Genéticamente , Humanos , Inmunidad Innata , Tropismo Viral
13.
J Immunol ; 195(10): 4933-42, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26466955

RESUMEN

Compartmentalization of nucleic acid (NA)-sensing TLR3, 7, 8, and 9 is strictly regulated to direct optimal response against microbial infection and evade recognition of host-derived NAs. Uncoordinated 93 homolog B1 (UNC93B1) is indispensable for trafficking of NA-sensing TLRs from the endoplasmic reticulum (ER) to endosomes/lysosomes. UNC93B1 controls loading of the TLRs into COPII vesicles to exit from the ER and traffics with the TLRs in the steady state. Ligand-induced translocation also happens on NA-sensing TLRs. However, the molecular mechanism for ligand-dependent trafficking of TLRs from the ER to endosomes/lysosomes remains unclear. In this study, we demonstrated that leucine-rich repeat containing protein (LRRC) 59, an ER membrane protein, participated in trafficking of NA-sensing TLRs from the ER. Knockdown of LRRC59 reduced TLR3-, 8-, and 9-mediated, but not TLR4-mediated, signaling. Upon ligand stimulation, LRRC59 associated with UNC93B1 in a TLR-independent manner, which required signals induced by ligand internalization. Endosomal localization of endogenous TLR3 was decreased by silencing of LRRC59, suggesting that LRRC59 promotes UNC93B1-mediated translocation of NA-sensing TLRs from the ER upon infection. These findings help us understand how NA-sensing TLRs control their proper distribution in the infection/inflammatory state.


Asunto(s)
Retículo Endoplásmico/inmunología , Infecciones/inmunología , Proteínas de la Membrana/inmunología , Proteínas de Transporte de Membrana/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Retículo Endoplásmico/genética , Endosomas/genética , Endosomas/inmunología , Células HEK293 , Humanos , Infecciones/genética , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Transducción de Señal/genética , Receptores Toll-Like/genética
14.
J Immunol ; 195(9): 4456-65, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26408662

RESUMEN

TLR4 triggers LPS signaling through the adaptors Toll/IL-1R domain-containing adaptor molecule (TICAM)-2 (also called TRAM) and TICAM-1 (also called TRIF), together with Toll/IL-1R domain-containing adaptor protein (TIRAP) and MyD88. The MyD88 pathway mediates early phase responses to LPS on the plasma membrane, whereas the TICAM pathway mediates late-phase responses, which induce the production of type I IFN and activation of inflammasomes. TICAM-2 bridges TLR4 and TICAM-1 for LPS signaling in the endosome. Recently, we identified an acidic motif, E87/D88/D89 in TICAM-2, that provides the interaction surfaces between TICAM-2 and TICAM-1. In the present study, we found additional D91/E92 in TICAM-2, conserved across species, that is crucial for TICAM-1 activation. The D91A/E92A mutant protein was distributed largely to the cytosol, despite myristoylation, suggesting its importance for assistance of membrane localization of TICAM-2. An ectopically expressed D91A/E92A mutant per se failed to activate TICAM-1, unlike its wild-type counterpart that forms self-aggregation, but it still retained the ability to pass LPS-mediated IFN regulatory factor (IRF)3 activation. In a TICAM-2 knockout human cell line expressing TLR4/MD-2 with or without CD14, overexpression of the D91A/E92A mutant did not activate IRF3, but upon LPS stimulation, it induced sufficient TLR4-mediated IRF3 activation with high coefficient colocalization. Hence, the D91/E92 motif guides TICAM-2 membrane localization and self-activation for signaling. Our results suggest the presence of two distinct steps underlying endosomal LPS signaling on TICAM-2 for TICAM-1 activation: TICAM-2 assembling in TLR4 and/or TICAM-2 self-activation. D91A/E92A of TICAM-2 selectively associates the TLR4-dependent TICAM-2 assembling, but not cytosolic TICAM-2 self-aggregation, to activate TICAM-1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Membrana Celular/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Endosomas/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/metabolismo , Microscopía Confocal , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
15.
Cancer Sci ; 107(5): 644-52, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26931406

RESUMEN

Transforming growth factor-ß activated kinase 1 (TAK1) has been shown to play a crucial role in cell death, differentiation, and inflammation. Here, we live-imaged robust TAK1 activation in Lewis lung carcinoma 3LL cells implanted into the s.c. tissue of syngeneic C57BL/6 mice and treated with polyinosinic:polycytidylic acid (PolyI:C). First, we developed and characterized a Förster resonance energy transfer-based biosensor for TAK1 activity. The TAK1 biosensor, named Eevee-TAK1, responded to stress-inducing reagents such as anisomycin, tumor necrosis factor-α, and interleukin1-ß. The anisomycin-induced increase in Förster resonance energy transfer was abolished by the TAK1 inhibitor (5z)-7-oxozeaenol. Activity of TAK1 in 3LL cells was markedly increased by PolyI:C in the presence of macrophages. 3LL cells expressing Eevee-TAK1 were implanted into mice and observed through imaging window by two-photon excitation microscopy. During the growth of tumor, the 3LL cells at the periphery of the tumor showed higher TAK1 activity than the 3LL cells located at the center of the tumor, suggesting that cells at the periphery of the tumor mass were under stronger stress. Injection of PolyI:C, which is known to induce regression of the implanted tumors, induced marked and homogenous TAK1 activation within the tumor tissues. The effect of PolyI:C faded within 4 days. These observations suggest that Eevee-TAK1 is a versatile tool to monitor cellular stress in cancer tissues.


Asunto(s)
Técnicas Biosensibles/métodos , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/enzimología , Quinasas Quinasa Quinasa PAM/metabolismo , Imagen Molecular/métodos , Poli I-C/uso terapéutico , Animales , Anisomicina/farmacología , Carcinoma Pulmonar de Lewis/patología , Supervivencia Celular , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía , Poli I-C/farmacología , Estrés Fisiológico/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Zearalenona/análogos & derivados , Zearalenona/farmacología
16.
BMC Immunol ; 17(1): 9, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-27141827

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Macrófagos/inmunología , Receptores Inmunológicos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Inmunidad Innata , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dominios Proteicos/genética , ARN Bicatenario/inmunología , Receptor de Interferón alfa y beta/genética , Receptores Inmunológicos/genética
17.
Biochem Biophys Res Commun ; 478(4): 1764-71, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27608599

RESUMEN

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.


Asunto(s)
Proliferación Celular/genética , Células Asesinas Naturales/metabolismo , Melanoma Experimental/genética , Proteínas de la Membrana/genética , Carga Tumoral/genética , Animales , Western Blotting , Línea Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Interferón beta/genética , Interferón beta/metabolismo , Interleucina-33/genética , Interleucina-33/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Microbiol Immunol ; 60(7): 511-5, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27240729

RESUMEN

Eye spray influenza vaccines for chickens are increasingly available; however, how to enhance cellular and antibody responses to them remains undetermined. Here, eye-drops containing the immune-enhancing adjuvants Pam2CSK4 or polyI:C were assessed in chickens. Application of these TLR agonists to chicken conjunctiva resulted in up-regulation of IL-1ß, but not other cytokines, including IFN and IL-6, in the spleen, lung and Harderian gland. Thus, responses to adjuvant applied to the conjunctival mucosa of chickens differ from those expected from the responses to intra-nasal adjuvants in mammals. Identifying an appropriate delivery route for adjuvants is crucial for evoking immune responses in chickens.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Pollos/inmunología , Pollos/metabolismo , Citocinas/biosíntesis , Inmunidad , Vacunas/inmunología , Animales , Anticuerpos Antivirales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Ligandos , Lipopéptidos/administración & dosificación , Masculino , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Poli I-C/administración & dosificación
19.
J Immunol ; 193(10): 5118-28, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25297876

RESUMEN

Nucleic acid-sensing TLRs are involved in both antimicrobial immune responses and autoimmune inflammation. TLR8 is phylogenetically and structurally related to TLR7 and TLR9, which undergo proteolytic processing in the endolysosomes to generate functional receptors. Recent structural analyses of human TLR8 ectodomain and its liganded form demonstrated that TLR8 is also cleaved, and both the N- and C-terminal halves contribute to ligand binding. However, the structures and ssRNA recognition mode of endogenous TLR8 in human primary cells are largely unknown. In this study, we show that proteolytic processing of TLR8 occurs in human monocytes and macrophages in a different manner compared with TLR7/9 cleavage. The insertion loop between leucine-rich repeats 14 and 15 in TLR8 is indispensable for the cleavage and stepwise processing that occurs in the N-terminal fragment. Both furin-like proprotein convertase and cathepsins contribute to TLR8 cleavage in the early/late endosomes. TLR8 recognizes viral ssRNA and endogenous RNA, such as microRNAs, resulting in the production of proinflammatory cytokines. Hence, localization sites of the receptors are crucial for the nucleic acid-sensing mode and downstream signaling.


Asunto(s)
Endosomas/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , ARN Viral/metabolismo , Receptor Toll-Like 8/metabolismo , Secuencia de Aminoácidos , Catepsinas/metabolismo , Expresión Génica , Células HEK293 , Humanos , Ligandos , Lisosomas/metabolismo , Macrófagos/química , Macrófagos/citología , MicroARNs/química , MicroARNs/metabolismo , Datos de Secuencia Molecular , Monocitos/química , Monocitos/citología , Cultivo Primario de Células , Proproteína Convertasa 1/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteolisis , ARN Viral/química , Transducción de Señal , Receptor Toll-Like 7/química , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/química , Receptor Toll-Like 8/genética , Receptor Toll-Like 9/química , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
20.
J Immunol ; 192(6): 2770-7, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24532585

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatocitos/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígenos CD8/inmunología , Antígenos CD8/metabolismo , Línea Celular , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/virología , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Hepacivirus/genética , Hepacivirus/fisiología , Hepatitis C/genética , Hepatitis C/metabolismo , Hepatocitos/metabolismo , Hepatocitos/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/inmunología , Poli I-C/farmacología , ARN Viral/genética , ARN Viral/inmunología , ARN Viral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA