Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Cell Rep ; 43(4): 113981, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38520688

RESUMO

Cholera toxin (CT), a bacterial exotoxin composed of one A subunit (CTA) and five B subunits (CTB), functions as an immune adjuvant. CTB can induce production of interleukin-1ß (IL-1ß), a proinflammatory cytokine, in synergy with a lipopolysaccharide (LPS), from resident peritoneal macrophages (RPMs) through the pyrin and NLRP3 inflammasomes. However, how CTB or CT activates these inflammasomes in the macrophages has been unclear. Here, we clarify the roles of inositol-requiring enzyme 1 alpha (IRE1α), an endoplasmic reticulum (ER) stress sensor, in CT-induced IL-1ß production in RPMs. In RPMs, CTB is incorporated into the ER and induces ER stress responses, depending on GM1, a cell membrane ganglioside. IRE1α-deficient RPMs show a significant impairment of CT- or CTB-induced IL-1ß production, indicating that IRE1α is required for CT- or CTB-induced IL-1ß production in RPMs. This study demonstrates the critical roles of IRE1α in activation of both NLRP3 and pyrin inflammasomes in tissue-resident macrophages.


Assuntos
Toxina da Cólera , Estresse do Retículo Endoplasmático , Endorribonucleases , Interleucina-1beta , Proteínas Serina-Treonina Quinases , Interleucina-1beta/metabolismo , Animais , Endorribonucleases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Camundongos , Toxina da Cólera/farmacologia , Toxina da Cólera/metabolismo , Inflamassomos/metabolismo , Camundongos Endogâmicos C57BL , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Lipopolissacarídeos/farmacologia , Retículo Endoplasmático/metabolismo
2.
J Exp Med ; 220(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37462944

RESUMO

Loss-of-function mutations in the lysosomal nucleoside transporter SLC29A3 cause lysosomal nucleoside storage and histiocytosis: phagocyte accumulation in multiple organs. However, little is known about the mechanism by which lysosomal nucleoside storage drives histiocytosis. Herein, histiocytosis in Slc29a3-/- mice was shown to depend on Toll-like receptor 7 (TLR7), which senses a combination of nucleosides and oligoribonucleotides (ORNs). TLR7 increased phagocyte numbers by driving the proliferation of Ly6Chi immature monocytes and their maturation into Ly6Clow phagocytes in Slc29a3-/- mice. Downstream of TLR7, FcRγ and DAP10 were required for monocyte proliferation. Histiocytosis is accompanied by inflammation in SLC29A3 disorders. However, TLR7 in nucleoside-laden splenic monocytes failed to activate inflammatory responses. Enhanced production of proinflammatory cytokines was observed only after stimulation with ssRNAs, which would increase lysosomal ORNs. Patient-derived monocytes harboring the G208R SLC29A3 mutation showed enhanced survival and proliferation in a TLR8-antagonist-sensitive manner. These results demonstrated that TLR7/8 responses to lysosomal nucleoside stress drive SLC29A3 disorders.


Assuntos
Histiocitose , Receptor 7 Toll-Like , Animais , Camundongos , Citocinas/genética , Histiocitose/genética , Mutação/genética , Nucleosídeos , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética
3.
Proc Natl Acad Sci U S A ; 119(45): e2121989119, 2022 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-36322717

RESUMO

Persistent mechanical pain hypersensitivity associated with peripheral inflammation, surgery, trauma, and nerve injury impairs patients' quality of life and daily activity. However, the molecular mechanism and treatment are not yet fully understood. Herein, we show that chemical ablation of isolectin B4-binding (IB4+) afferents by IB4-saporin injection into sciatic nerves completely and selectively inhibited inflammation- and tissue injury-induced mechanical pain hypersensitivity while thermal and mechanical pain hypersensitivities were normal following nerve injury. To determine the molecular mechanism involving the specific types of mechanical pain hypersensitivity, we compared gene expression profiles between IB4+ neuron-ablated and control dorsal root ganglion (DRG) neurons. We identified Tmem45b as one of 12 candidate genes that were specific to somatosensory ganglia and down-regulated by IB4+ neuronal ablation. Indeed, Tmem45b was expressed predominantly in IB4+ DRG neurons, where it was selectively localized in the trans Golgi apparatus of DRG neurons but not detectable in the peripheral and central branches of DRG axons. Tmem45b expression was barely detected in the spinal cord and brain. Although Tmem45b-knockout mice showed normal responses to noxious heat and noxious mechanical stimuli under normal conditions, mechanical pain hypersensitivity was selectively impaired after inflammation and tissue incision, reproducing the pain phenotype of IB4+ sensory neuron-ablated mice. Furthermore, acute knockdown by intrathecal injection of Tmem45b small interfering RNA, either before or after inflammation induction, successfully reduced mechanical pain hypersensitivity. Thus, our study demonstrates that Tmem45b is essential for inflammation- and tissue injury-induced mechanical pain hypersensitivity and highlights Tmem45b as a therapeutic target for future treatment.


Assuntos
Hipersensibilidade , Qualidade de Vida , Animais , Camundongos , Gânglios Espinais/metabolismo , Hipersensibilidade/metabolismo , Inflamação/metabolismo , Camundongos Knockout , Dor/genética , Dor/complicações , Células Receptoras Sensoriais/metabolismo
4.
Vet Immunol Immunopathol ; 253: 110505, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36327941

RESUMO

Cancers utilize a variety of molecules to escape host immune responses. Better understanding the immune environment surrounding cancer may facilitate application of innovative cancer immunotherapies, such as immune checkpoint inhibitors, to dogs as well as humans. In this study, we screened the expression of 20 immune regulatory molecules in diverse canine tumors (n = 59). Quantitative RT-PCR (qPCR) analysis revealed that some immune regulatory molecules, such as LGALS9 (coding Galectin-9) and CD48, were expressed in most canine tumors, but other molecules, such as CD274 (coding PD-L1), IL4I1, PVR, TNFSF18, ICOSLG, and TNFSF4, were rarely expressed. NECTIN2 was highly expressed in epithelial tumors but was low in non-epithelial tumors. In contrast, VSIR and CD200 expressions were low in epithelial tumors but high in non-epithelial tumors. Interestingly, several tumors expressed distinctive immunoregulatory factors. Hepatocellular carcinomas expressed FGL1, mast cell tumors expressed PDCD1LG2 (coding PD-L2), transitional cell carcinomas expressed VTCN1 (coding B7x), and lymphomas and squamous cell carcinomas expressed CD70. Consistent with qPCR results, immunofluorescence staining confirmed that hepatocellular carcinomas expressed FGL-1 protein. Thus, this study reveals the expression profile of immunoregulatory molecules in canine tumors and opens the door to better understanding the relationship between canine tumors and host immunity.


Assuntos
Carcinoma Hepatocelular , Carcinoma de Células Escamosas , Doenças do Cão , Neoplasias Hepáticas , Animais , Cães , Antígeno B7-H1 , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/veterinária , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/veterinária , Fibrinogênio , Imunidade , L-Aminoácido Oxidase , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/veterinária , Ligante OX40
5.
Front Immunol ; 13: 857954, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35693801

RESUMO

Dendritic cells (DC) play critical roles in linking innate and adaptive immunity. DC are heterogenous and there are subsets with various distinct functions. One DC subset, conventional type 1 DC (cDC1), can be defined by expression of CD8α/CD103 in mice and CD141 in humans, or by expression of a chemokine receptor, XCR1, which is a conserved marker in both mice and human. cDC1 are characterized by high ability to ingest dying cells and to cross-present antigens for generating cytotoxic CD8 T cell responses. Through these activities, cDC1 play crucial roles in immune responses against infectious pathogens or tumors. Meanwhile, cDC1 involvement in homeostatic situations is not fully understood. Analyses by using mutant mice, in which cDC1 are ablated in vivo, revealed that cDC1 are critical for maintaining intestinal immune homeostasis. Here, we review the homeostatic roles of cDC1, focusing upon intestinal immunity.


Assuntos
Apresentação Cruzada , Células Dendríticas , Animais , Linfócitos T CD8-Positivos , Homeostase , Camundongos , Receptores de Quimiocinas/metabolismo
6.
Nat Commun ; 12(1): 6819, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34819510

RESUMO

Impaired proteasome activity due to genetic variants of certain subunits might lead to proteasome-associated autoinflammatory syndromes (PRAAS). Here we report a de novo heterozygous missense variant of the PSMB9 proteasome subunit gene in two unrelated Japanese infants resulting in amino acid substitution of the glycine (G) by aspartic acid (D) at position 156 of the encoded protein ß1i. In addition to PRAAS-like manifestations, these individuals suffer from pulmonary hypertension and immunodeficiency, which are distinct from typical PRAAS symptoms. The missense variant results in impaired immunoproteasome maturation and activity, yet ubiquitin accumulation is hardly detectable in the patients. A mouse model of the heterozygous human genetic variant (Psmb9G156D/+) recapitulates the proteasome defects and the immunodeficiency phenotype of patients. Structurally, PSMB9 G156D interferes with the ß-ring-ßring interaction of the wild type protein that is necessary for 20S proteasome formation. We propose the term, proteasome-associated autoinflammatory syndrome with immunodeficiency (PRAAS-ID), to indicate a separate category of autoinflammatory diseases, similar to, but distinct from PRAAS, that describes the patients in this study.


Assuntos
Cisteína Endopeptidases/genética , Doenças Hereditárias Autoinflamatórias/genética , Hipertensão Pulmonar/genética , Doenças da Imunodeficiência Primária/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Cisteína Endopeptidases/metabolismo , Modelos Animais de Doenças , Feminino , Doenças Hereditárias Autoinflamatórias/diagnóstico , Doenças Hereditárias Autoinflamatórias/imunologia , Doenças Hereditárias Autoinflamatórias/patologia , Heterozigoto , Humanos , Hipertensão Pulmonar/diagnóstico , Hipertensão Pulmonar/imunologia , Recém-Nascido , Masculino , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Linhagem , Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/imunologia , Doenças da Imunodeficiência Primária/patologia , Complexo de Endopeptidases do Proteassoma/genética , Síndrome
7.
Arthritis Rheumatol ; 73(11): 2105-2115, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33982886

RESUMO

OBJECTIVE: Coatomer subunit alpha (COPA) syndrome, also known as autoinflammatory interstitial lung, joint, and kidney disease, is caused by heterozygous mutations in COPA. We identified a novel COPA variant in 4 patients in one family. We undertook this study to elucidate whether and how the variant causes manifestations of COPA syndrome by studying these 4 patients and by analyzing results from a gene-targeted mouse model. METHODS: We performed whole-exome sequencing in 7 family members and measured the type I interferon (IFN) signature of the peripheral blood cells. We analyzed the effects of COPA variants in in vitro experiments and in Copa mutant mice that were generated. RESULTS: We identified a heterozygous variant of COPA (c.725T>G, p.Val242Gly) in the 4 affected members of the family. The IFN score was high in the members carrying the variant. In vitro analysis revealed that COPA V242G, as well as the previously reported disease-causing variants, augmented stimulator of interferon genes (STING)-induced type I IFN promoter activities. CopaV242G/+ mice manifested interstitial lung disease and STING-dependent elevation of IFN-stimulated gene expression. In CopaV242G/+ dendritic cells, the STING pathway was not constitutively activated but was hyperactivated upon stimulation, leading to increased type I IFN production. CONCLUSION: V242G, a novel COPA variant, was found in 4 patients from one family. In gene-targeted mice with the V242G variant, interstitial lung disease was recapitulated and augmented responses of the STING pathway, leading to an increase in type I IFN production, were demonstrated.


Assuntos
Proteína Coatomer/genética , Interferon Tipo I/genética , Artropatias/genética , Nefropatias/genética , Doenças Pulmonares Intersticiais/genética , Mutação de Sentido Incorreto , Alelos , Análise Mutacional de DNA , Feminino , Heterozigoto , Humanos , Artropatias/imunologia , Nefropatias/imunologia , Doenças Pulmonares Intersticiais/imunologia , Masculino , Linhagem , Sequenciamento do Exoma
8.
Sci Rep ; 11(1): 10439, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34001954

RESUMO

The thymus facilitates mature T cell production by providing a suitable stromal microenvironment. This microenvironment is impaired by radiation and aging which lead to immune system disturbances known as thymic involution. Young adult thymus shows thymic recovery after such involution. Although various genes have been reported for thymocytes and thymic epithelial cells in such processes, the roles of stromal transcription factors in these remain incompletely understood. MafB (v-maf musculoaponeurotic fibrosarcoma oncogene homolog B) is a transcription factor expressed in thymic stroma and its expression was induced a day after radiation exposure. Hence, the roles of mesenchymal MafB in the process of thymic regeneration offers an intriguing research topic also for radiation biology. The current study investigated whether MafB plays roles in the adult thymus. MafB/green fluorescent protein knock-in mutant (MafB+/GFP) mice showed impaired thymic regeneration after the sublethal irradiation, judged by reduced thymus size, total thymocyte number and medullary complexity. Furthermore, IL4 was induced after irradiation and such induction was reduced in mutant mice. The mutants also displayed signs of accelerated age-related thymic involution. Altogether, these results suggest possible functions of MafB in the processes of thymic recovery after irradiation, and maintenance during aging.


Assuntos
Fator de Transcrição MafB/metabolismo , Regeneração/efeitos da radiação , Timócitos/fisiologia , Timo/fisiologia , Envelhecimento/genética , Animais , Proliferação de Células/genética , Proliferação de Células/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Técnicas de Introdução de Genes , Fator de Transcrição MafB/genética , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Regeneração/genética , Timócitos/efeitos da radiação , Timo/citologia , Timo/efeitos da radiação , Irradiação Corporal Total
9.
iScience ; 24(5): 102424, 2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-33997702

RESUMO

Immunogenic tumor cell death enhances anti-tumor immunity. However, the mechanisms underlying this effect are incompletely understood. We established a system to induce tumor cell death in situ and investigated its effect on dendritic cell (DC) migration and T cell responses using intravital photolabeling in mice expressing KikGR photoconvertible protein. We demonstrate that tumor cell death induces phagocytosis of tumor cells by tumor-infiltrating (Ti)-DCs, and HMGB1-TLR4 and ATP-P2X7 receptor signaling-dependent Ti-DC emigration to draining lymph nodes (dLNs). This led to an increase in anti-tumor CD8+ T cells of memory precursor effector phenotype and secondary tumor growth inhibition in a CD103+ DC-dependent manner. However, combining tumor cell death induction with lipopolysaccharide treatment stimulated Ti-DC maturation and emigration to dLNs but did not improve tumor immunity. Thus, immunogenic tumor cell death enhances tumor immunity by increasing Ti-DC migration to dLNs where they promote anti-tumor T cell responses and tumor growth inhibition.

10.
Biochem Biophys Res Commun ; 525(2): 477-482, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32111355

RESUMO

Plasmacytoid dendritic cells (pDCs) are characterized by an exclusive expression of nucleic acid sensing Toll-like receptor 7 (TLR7) and TLR9, and production of high amounts of type I interferon (IFN) in response to TLR7/9 signaling. This function is crucial for both antiviral immunity and the pathogenesis of autoimmune diseases. An Ets family transcription factor, i.e., Spi-B (which is highly expressed in pDCs) is required for TLR7/9 signal-induced type I IFN production and can transactivate IFN-α promoter in synergy with IFN regulatory factor-7 (IRF-7). Herein, we analyzed how Spi-B contributes to the transactivation of the Ifna4 promoter. We performed deletion and/or mutational analyses of the Ifna4 promoter and an electrophoretic mobility shift assay (EMSA) and observed an Spi-B binding site in close proximity to the IRF-7 binding site. The EMSA results also showed that the binding of Spi-B to the double-stranded DNA probe potentiated the recruitment of IRF-7 to its binding site. We also observed that the association of Spi-B with transcriptional coactivator p300 was required for the Spi-B-induced synergistic enhancement of the Ifna4 promoter activity by Spi-B. These results clarify the molecular mechanism of action of Spi-B in the transcriptional activation of the Ifna4 promoter.


Assuntos
Interferon-alfa/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Ativação Transcricional , Animais , Proteína p300 Associada a E1A/metabolismo , Células HEK293 , Humanos , Camundongos , Mutação , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-ets/genética
11.
Br J Cancer ; 122(8): 1185-1193, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32066911

RESUMO

BACKGROUND: Cancer peptide vaccines show only marginal effects against cancers. Immune checkpoint inhibitors (ICIs) show significant curative effects in certain types of cancers, but the response rate is still limited. In this study, we aim to improve cancer peptide vaccination by targeting Ag peptides selectively to a dendritic cell (DC) subset, XCR1-expressing DCs (XCR1+ DCs), with high ability to support CD8+ T-cell responses. METHODS: We have generated a fusion protein, consisting of an Ag peptide presented with MHC class I, and an XCR1 ligand, XCL1, and examined its effects on antitumour immunity in mice. RESULTS: The fusion protein was delivered to XCR1+ DCs in an XCR1-dependent manner. Immunisation with the fusion protein plus an immune adjuvant, polyinosinic:polycytidylic acids (poly(I:C)), more potently induced Ag-specific CD8+ T-cell responses through XCR1 than the Ag peptide plus poly(I:C) or the Ag protein plus poly(I:C). The fusion protein plus poly(I:C) inhibited the tumour growth efficiently in the prophylactic and therapeutic tumour models. Furthermore, the fusion protein plus poly(I:C) showed suppressive effects on tumour growth in synergy with anti-PD-1 Ab. CONCLUSIONS: Cancer Ag targeting to XCR1+ DCs should be a promising procedure as a combination anticancer therapy with immune checkpoint blockade.


Assuntos
Antígenos/imunologia , Vacinas Anticâncer/imunologia , Quimiocinas C/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/terapia , Poli I-C/farmacologia , Vacinas de Subunidades Antigênicas/imunologia
12.
Int J Hematol ; 109(6): 744-750, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30706328

RESUMO

Aggressive natural killer cell leukemia (ANKL) is a rare neoplasm characterized by the systemic infiltration of Epstein-Barr virus (EBV)-associated NK cells, and rapidly progressive clinical course. We report the case of a 45-year-old man with intellectual disability who developed ANKL, and describe the identification of a novel genetic mutation of coiled-coil domain-containing 22 (CCDC22). He presented with persistent fever, severe pancytopenia, and hepatosplenomegary. Following bone marrow aspiration, numerous hemophagocytes were identified. High EBV viral load was detected in NK cells fractionation by qPCR. The initial diagnosis was EBV-related hemophagocytic lymphohistiocytosis (EBV-HLH). A combination of immunosuppressive drugs and chemotherapy was administered, but was unsuccessful in controlling the disease. Therefore, he was treated with HLA-matched related allogeneic hematopoietic stem cell transplantation. However, his condition deteriorated within 30 days, resulting in fatal outcome. Autopsy revealed many EBV-infected NK cells infiltrating major organs, consistent with ANKL. Furthermore, whole-exome sequencing identified a novel missense mutation of the CCDC22 gene (c.112G>A, p.V38M), responsible for X-linked intellectual disability (XLID). CCDC22 has been shown to play a role in NF-κB activation. Our case suggests that CCDC22 mutation might be implicated in pathogenesis of EBV-HLH and NK-cell neoplasms as well as XLID via possibly affecting NF-κB signaling.


Assuntos
Infecções por Vírus Epstein-Barr/genética , Leucemia Linfocítica Granular Grande/genética , Linfo-Histiocitose Hemofagocítica/genética , Mutação de Sentido Incorreto , Proteínas/genética , Aloenxertos , Cromossomos Humanos X/genética , Transplante de Células-Tronco Hematopoéticas , Humanos , Deficiência Intelectual/genética , Leucemia Linfocítica Granular Grande/terapia , Linfo-Histiocitose Hemofagocítica/terapia , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Índice de Gravidade de Doença , Transdução de Sinais/genética
13.
Cancer Sci ; 110(4): 1279-1292, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30702189

RESUMO

Programmed cell death ligand 1 (PD-L1) on tumor cells suppresses anti-tumor immunity and has an unfavorable prognostic impact in ovarian cancer patients. We herein report the pathophysiological and therapeutic impacts of PD-L1 disruption in ovarian cancer. PD-L1 was genetically disrupted in the murine ovarian cancer cell line ID8 using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-mediated genome editing. PD-L1 knockout (KO) and control ovarian cancer cells were intraperitoneally inoculated into syngeneic mice, and survival and tumor dissemination were evaluated. Survival times were significantly longer in the PD-L1-KO ID8-inoculated groups than in their control groups, and its therapeutic benefit was enhanced in combination with the cisplatin treatment. Tumor weights and ascites volumes were significantly lower in the PD-L1-KO ID8 groups than in their control groups. Immunohistochemical and immunofluorescence analyses showed that intratumoral CD4+ T cells, CD8+ T cells, NK cells and CD11c+ M1 macrophages were significantly increased, whereas regulatory T cells were significantly decreased in the PD-L1-KO ID8 groups compared with those in their control groups. The intratumoral mRNA expression of interferon-γ, tumor-necrosis factor-α, interleukin (IL)-2, IL-12a, CXCL9 and CXCL10 was significantly stronger, while that of IL-10, vascular endothelial growth factor, CXCL1 and CXCL2 was significantly weaker in the PD-L1-KO ID8 groups. These results indicate that CRISPR/Cas9-mediated PD-L1 disruption on tumor cells promotes anti-tumor immunity by increasing tumor-infiltrating lymphocytes and modulating cytokine/chemokine profiles within the tumor microenvironment, thereby suppressing ovarian cancer progression. These results suggest that PD-L1-targeted therapy by genome editing may be a novel therapeutic strategy for ovarian cancer.


Assuntos
Antígeno B7-H1/metabolismo , Sistemas CRISPR-Cas , Edição de Genes , Imunidade , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Animais , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Citocinas/metabolismo , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Deleção de Genes , Loci Gênicos , Humanos , Imunomodulação , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Metástase Neoplásica , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia
14.
J Invest Dermatol ; 139(6): 1329-1338.e7, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30776434

RESUMO

Chronic low-grade inflammation can cause several metabolic syndromes. Patients with psoriasis, a chronic immunological skin inflammation, often develop diabetes. However, it is not clear to date how psoriasis leads to, or is correlated with, glucose intolerance. Here, we investigate whether psoriasis itself is correlated with hyperglycemia in humans and mice. In patients, the severity of psoriasis was correlated with high blood glucose levels, and treatment of psoriasis by phototherapy improved insulin secretion. Imiquimod-induced systemic and cutaneous inflammation in mice, with features of human psoriasis, also resulted in hyperglycemia. Although it should be determined if psoriasis-like cutaneous inflammation alone can induce hyperglycemia, imiquimod-treated mice showed impairment of insulin secretion without significant islet inflammation. Administration of anti-IL-17A monoclonal antibody improved hyperglycemia in patients with psoriasis and imiquimod-treated mice with psoriasiform features. These results suggest that hyperglycemia is highly associated with psoriasis, mainly through IL-17.


Assuntos
Hiperglicemia/epidemiologia , Interleucina-17/imunologia , Psoríase/complicações , Animais , Glicemia/análise , Estudos Transversais , Modelos Animais de Doenças , Feminino , Hemoglobinas Glicadas/análise , Humanos , Hiperglicemia/sangue , Hiperglicemia/diagnóstico , Hiperglicemia/imunologia , Imiquimode/imunologia , Insulina/metabolismo , Interleucina-17/antagonistas & inibidores , Masculino , Camundongos , Pessoa de Meia-Idade , Fototerapia , Psoríase/diagnóstico , Psoríase/imunologia , Psoríase/terapia , Estudos Retrospectivos , Índice de Gravidade de Doença , Pele , Resultado do Tratamento
15.
Int Immunol ; 31(10): 657-668, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30689886

RESUMO

Cholera toxin B (CTB) is a subunit of cholera toxin, a bacterial enterotoxin secreted by Vibrio cholerae and also functions as an immune adjuvant. However, it remains unclear how CTB activates immune cells. We here evaluated whether or how CTB induces production of a pro-inflammatory cytokine, interleukin-1ß (IL-1ß). CTB induced IL-1ß production not only from bone marrow-derived macrophages (BMMs) but also from resident peritoneal macrophages in synergy with O111:B4-derived lipopolysaccharide (LPS O111:B4) that can bind to CTB. Meanwhile, when prestimulated with O55:B5-derived LPS (LPS O55:B5) that fails to bind to CTB, resident peritoneal macrophages, but not BMMs, produced IL-1ß in response to CTB. The CTB-induced IL-1ß production in synergy with LPS in both peritoneal macrophages and BMMs was dependent on ganglioside GM1, which is required for internalization of CTB. Notably, not only the NLRP3 inflammasome but also the pyrin inflammasome were involved in CTB-induced IL-1ß production from resident peritoneal macrophages, while only the NLRP3 inflammasome was involved in that from BMMs. In response to CTB, a Rho family small GTPase, RhoA, which activates pyrin inflammasome upon various kinds of biochemical modification, increased its phosphorylation at serine-188 in a GM1-dependent manner. This phosphorylation as well as CTB-induced IL-1ß productions were dependent on protein kinase A (PKA), indicating critical involvement of PKA-dependent RhoA phosphorylation in CTB-induced IL-1ß production. Taken together, these results suggest that CTB, incorporated through GM1, can activate resident peritoneal macrophages to produce IL-1ß in synergy with LPS through novel mechanisms in which pyrin as well as NLRP3 inflammasomes are involved.


Assuntos
Toxina da Cólera/farmacologia , Inflamassomos/efeitos dos fármacos , Interleucina-1beta/biossíntese , Macrófagos Peritoneais/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Pirina/imunologia , Animais , Humanos , Inflamassomos/imunologia , Macrófagos Peritoneais/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
16.
Gan To Kagaku Ryoho ; 45(10): 1469-1471, 2018 Oct.
Artigo em Japonês | MEDLINE | ID: mdl-30382048

RESUMO

Tumor-derived peptides can induce antitumor cytotoxic T lymphocyte(CTL)response. However, the effects are limited. We aimed to overcome this limitation by selectively delivering antigen peptides to an XC chemokine receptor 1-expressing dendritic cell subset(XCR1+DC)that is notable for its exceptional ability to generate CTL response. To do that, we designed a vaccine(mXCL1-OVA peptide vaccine)that consisted of a murine XCR1 ligand(XCL1)and an ovalbumin(OVA)-derived MHC class I-restricted antigen. When co-injected with the immune adjuvant polyinosinic-polycytidylic acid(poly[I: C]), mXCL1-OVA peptide vaccine showed much greater antigen-specific cytotoxic T cell(CTL)response than either OVA protein plus poly(I: C)or OVA peptide plus poly(I: C). Furthermore, mXCL1-OVA peptide vaccine plus poly(I: C)showed more prominent antitumor effects against OVA-expressing melanoma(B16-OVA)than other vaccines with regard to growth inhibition. Thus, our results suggest that chemokine-directed antigen delivery to DC subsets with high CTL-inducing ability is a promising method for generating effective antitumor immunity.


Assuntos
Antígenos/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Neoplasias/terapia , Animais , Vacinas Anticâncer/uso terapêutico , Camundongos , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico
17.
J Immunol ; 200(1): 119-129, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158419

RESUMO

Skin dendritic cells (DCs) are divided into several subsets with distinctive functions. This study shows a previously unappreciated role of dermal CD11b-type Langerin- DCs in maintaining immunological self-tolerance after UVB exposure. After UVB exposure, dermal CD11b-type Langerin- DCs upregulated surface CD86 expression, induced proliferation of Foxp3+ regulatory T (Treg) cells without exogenous Ags, and upregulated a set of genes associated with immunological tolerance. This Treg-expansion activity was significantly hampered by CD80/CD86 blockade in vivo. These results indicate that CD11b-type Langerin- DCs from the UVB-exposed skin are specialized to expand Treg cells in the skin, which suppress autoimmunity.


Assuntos
Células Dendríticas/imunologia , Pele/patologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Tolerância Imunológica/genética , Lectinas Tipo C/metabolismo , Ativação Linfocitária , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transcriptoma , Raios Ultravioleta/efeitos adversos
18.
Sci Rep ; 7(1): 16833, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203799

RESUMO

The potential role of macrophages in pulmonary fibrosis (PF) prompted us to evaluate the roles of CX3CR1, a chemokine receptor abundantly expressed in macrophages during bleomycin (BLM)-induced PF. Intratracheal BLM injection induced infiltration of leukocytes such as macrophages into the lungs, which eventually resulted in fibrosis. CX3CR1 expression was mainly detected in the majority of macrophages and in a small portion of α-smooth muscle actin-positive cells in the lungs, while CX3CL1 was expressed in macrophages. BLM-induced fibrotic changes in the lungs were reduced without any changes in the number of leukocytes in Cx3cr1 -/- mice, as compared with those in the wild-type (WT) mice. However, intrapulmonary CX3CR1+ macrophages displayed pro-fibrotic M2 phenotypes; lack of CX3CR1 skewed their phenotypes toward M1 in BLM-challenged lungs. Moreover, fibrocytes expressed CX3CR1, and were increased in BLM-challenged WT lungs. The number of intrapulmonary fibrocytes was decreased in Cx3cr1 -/- mice. Thus, locally-produced CX3CL1 can promote PF development primarily by attracting CX3CR1-expressing M2 macrophages and fibrocytes into the lungs.


Assuntos
Receptor 1 de Quimiocina CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Macrófagos/imunologia , Fibrose Pulmonar/patologia , Animais , Bleomicina/toxicidade , Líquido da Lavagem Broncoalveolar/citologia , Movimento Celular , Quimiocina CX3CL1/deficiência , Quimiocina CX3CL1/genética , Feminino , Leucócitos/citologia , Pulmão/patologia , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo
19.
Methods Mol Biol ; 1423: 247-53, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27142021

RESUMO

Dendritic cells (DCs) are one of the key populations controlling immune responses. To establish a cell depletion system in vivo, human diphtheria toxin (DT) receptor (DTR) is transduced to the mice in which DTR is expressed under the control of a specific promoter. In these mice, DTR-expressing cells are inducibly depleted after DT injection. Using this system, analysis of mouse models in which DTR was expressed under the CD11c promoter has contributed to our knowledge of DC biology by depleting CD11c(+) cells. Other mouse models to inducibly eliminate specific DC subsets upon DT treatment have been also generated. Here, we describe a new mouse model in which the XCR1(+) DC subset is inducibly and transiently depleted in vivo.


Assuntos
Antígeno CD11c/genética , Células Dendríticas/imunologia , Toxina Diftérica/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Técnicas de Introdução de Genes , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Humanos , Camundongos , Camundongos Transgênicos , Modelos Animais , Regiões Promotoras Genéticas , Receptores de Quimiocinas/genética
20.
Sci Rep ; 6: 23505, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27005831

RESUMO

Intestinal immune homeostasis requires dynamic crosstalk between innate and adaptive immune cells. Dendritic cells (DCs) exist as multiple phenotypically and functionally distinct sub-populations within tissues, where they initiate immune responses and promote homeostasis. In the gut, there exists a minor DC subset defined as CD103(+)CD11b(-) that also expresses the chemokine receptor XCR1. In other tissues, XCR1(+) DCs cross-present antigen and contribute to immunity against viruses and cancer, however the roles of XCR1(+) DCs and XCR1 in the intestine are unknown. We showed that mice lacking XCR1(+) DCs are specifically deficient in intraepithelial and lamina propria (LP) T cell populations, with remaining T cells exhibiting an atypical phenotype and being prone to death, and are also more susceptible to chemically-induced colitis. Mice deficient in either XCR1 or its ligand, XCL1, similarly possess diminished intestinal T cell populations, and an accumulation of XCR1(+) DCs in the gut. Combined with transcriptome and surface marker expression analysis, these observations lead us to hypothesise that T cell-derived XCL1 facilitates intestinal XCR1(+) DC activation and migration, and that XCR1(+) DCs in turn provide support for T cell survival and function. Thus XCR1(+) DCs and the XCR1/XCL1 chemokine axis have previously-unappreciated roles in intestinal immune homeostasis.


Assuntos
Quimiocinas C/metabolismo , Células Dendríticas/fisiologia , Intestinos/imunologia , Receptores de Quimiocinas/metabolismo , Linfócitos T/citologia , Animais , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Quimiocinas C/deficiência , Apresentação Cruzada , Células Dendríticas/imunologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Homeostase , Intestinos/citologia , Camundongos , Receptores de Quimiocinas/deficiência , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA