Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 6819, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819510

RESUMEN

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.


Asunto(s)
Cisteína Endopeptidasas/genética , Enfermedades Autoinflamatorias Hereditarias/genética , Hipertensión Pulmonar/genética , Enfermedades de Inmunodeficiencia Primaria/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Cisteína Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Femenino , Enfermedades Autoinflamatorias Hereditarias/diagnóstico , Enfermedades Autoinflamatorias Hereditarias/inmunología , Enfermedades Autoinflamatorias Hereditarias/patología , Heterocigoto , Humanos , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/inmunología , Recién Nacido , Masculino , Ratones , Ratones Transgénicos , Mutación Missense , Linaje , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/inmunología , Enfermedades de Inmunodeficiencia Primaria/patología , Complejo de la Endopetidasa Proteasomal/genética , Síndrome
2.
Arthritis Rheumatol ; 73(11): 2105-2115, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33982886

RESUMEN

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.


Asunto(s)
Proteína Coatómero/genética , Interferón Tipo I/genética , Artropatías/genética , Enfermedades Renales/genética , Enfermedades Pulmonares Intersticiales/genética , Mutación Missense , Alelos , Análisis Mutacional de ADN , Femenino , Heterocigoto , Humanos , Artropatías/inmunología , Enfermedades Renales/inmunología , Enfermedades Pulmonares Intersticiales/inmunología , Masculino , Linaje , Secuenciación del Exoma
3.
Br J Cancer ; 122(8): 1185-1193, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32066911

RESUMEN

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.


Asunto(s)
Antígenos/inmunología , Vacunas contra el Cáncer/inmunología , Quimiocinas C/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Ratones Endogámicos C57BL , Neoplasias Experimentales/terapia , Poli I-C/farmacología , Vacunas de Subunidad/inmunología
4.
Cancer Sci ; 110(4): 1279-1292, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30702189

RESUMEN

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.


Asunto(s)
Antígeno B7-H1/metabolismo , Sistemas CRISPR-Cas , Edición Génica , Inmunidad , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Animales , Antígeno B7-H1/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Citocinas/metabolismo , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Eliminación de Gen , Sitios Genéticos , Humanos , Inmunomodulación , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología
5.
Int Immunol ; 31(10): 657-668, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30689886

RESUMEN

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.


Asunto(s)
Toxina del Cólera/farmacología , Inflamasomas/efectos de los fármacos , Interleucina-1beta/biosíntesis , Macrófagos Peritoneales/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Pirina/inmunología , Animales , Humanos , Inflamasomas/inmunología , Macrófagos Peritoneales/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
6.
Sci Rep ; 6: 23505, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-27005831

RESUMEN

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.


Asunto(s)
Quimiocinas C/metabolismo , Células Dendríticas/fisiología , Intestinos/inmunología , Receptores de Quimiocina/metabolismo , Linfocitos T/citología , Animales , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Quimiocinas C/deficiencia , Reactividad Cruzada , Células Dendríticas/inmunología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Homeostasis , Intestinos/citología , Ratones , Receptores de Quimiocina/deficiencia , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...