Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
2.
Sci Immunol ; 6(60)2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145065

RESUMO

Analysis of autoinflammatory and immunodeficiency disorders elucidates human immunity and fosters the development of targeted therapies. Oligoadenylate synthetase 1 is a type I interferon-induced, intracellular double-stranded RNA (dsRNA) sensor that generates 2'-5'-oligoadenylate to activate ribonuclease L (RNase L) as a means of antiviral defense. We identified four de novo heterozygous OAS1 gain-of-function variants in six patients with a polymorphic autoinflammatory immunodeficiency characterized by recurrent fever, dermatitis, inflammatory bowel disease, pulmonary alveolar proteinosis, and hypogammaglobulinemia. To establish causality, we applied genetic, molecular dynamics simulation, biochemical, and cellular functional analyses in heterologous, autologous, and inducible pluripotent stem cell-derived macrophages and/or monocytes and B cells. We found that upon interferon-induced expression, OAS1 variant proteins displayed dsRNA-independent activity, which resulted in RNase L-mediated RNA cleavage, transcriptomic alteration, translational arrest, and dysfunction and apoptosis of monocytes, macrophages, and B cells. RNase L inhibition with curcumin modulated and allogeneic hematopoietic cell transplantation cured the disorder. Together, these data suggest that human OAS1 is a regulator of interferon-induced hyperinflammatory monocyte, macrophage, and B cell pathophysiology.


Assuntos
2',5'-Oligoadenilato Sintetase/genética , Doenças Hereditárias Autoinflamatórias/genética , Doenças da Imunodeficiência Primária/genética , 2',5'-Oligoadenilato Sintetase/imunologia , 2',5'-Oligoadenilato Sintetase/isolamento & purificação , 2',5'-Oligoadenilato Sintetase/metabolismo , Linfócitos B/imunologia , Células Cultivadas , Análise Mutacional de DNA , Endorribonucleases/genética , Endorribonucleases/metabolismo , Ensaios Enzimáticos , Mutação com Ganho de Função/imunologia , Técnicas de Inativação de Genes , Transplante de Células-Tronco Hematopoéticas , Doenças Hereditárias Autoinflamatórias/diagnóstico , Doenças Hereditárias Autoinflamatórias/imunologia , Doenças Hereditárias Autoinflamatórias/terapia , Heterozigoto , Humanos , Lactente , Recém-Nascido , Interferon Tipo I/metabolismo , Macrófagos/imunologia , Simulação de Dinâmica Molecular , Monócitos/imunologia , Cultura Primária de Células , Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/imunologia , Doenças da Imunodeficiência Primária/terapia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia
3.
J Invest Dermatol ; 141(3): 512-522, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32888953

RESUMO

α-(1,6)-fucosyltransferase 8 (FUT8) is implicated in the pathogenesis of several malignancies, but its role in psoriasis is poorly understood. In this study, we show that FUT8 remodeling of EGFR plays a critical role in the development of psoriasis phenotypes. Notably, elevated FUT8 expression was associated with disease severity in the lesional epidermis of a patient with psoriasis. FUT8 gain of function promoted HaCaT cell proliferation, whereas short hairpin FUT8 reduced cell proliferation and induced a longer S phase with downregulation of cyclin A1 expression. Furthermore, cell proliferation, which is controlled by the activation of EGFR, was shown to be regulated by FUT8 core fucosylation of EGFR. Short hairpin FUT8 significantly reduced EGFR/protein kinase B signaling and slowed EGF‒EGFR complex trafficking to the perinuclear region. Moreover, short hairpin FUT8 reduced ligand-induced EGFR dimerization. Overactivated EGFR was observed in the lesional epidermis of both human patient and psoriasis-like mouse model, whereas conditional knockout of FUT8 in an IL-23 psoriasis-like mouse model ameliorated disease phenotypes and reduced EGFR activation in the epidermis. These findings implied that elevated FUT8 expression in the lesional epidermis is implicated in the development of psoriasis phenotypes, being required for EGFR overactivation and leading to keratinocyte hyperproliferation.


Assuntos
Epiderme/patologia , Receptores ErbB/metabolismo , Fucosiltransferases/metabolismo , Psoríase/imunologia , Animais , Estudos de Casos e Controles , Proliferação de Células/genética , Modelos Animais de Doenças , Epiderme/imunologia , Feminino , Fucosiltransferases/genética , Mutação com Ganho de Função/imunologia , Glicosilação , Células HaCaT , Voluntários Saudáveis , Humanos , Interleucina-23/administração & dosagem , Interleucina-23/imunologia , Masculino , Camundongos Knockout , Cultura Primária de Células , Multimerização Proteica/imunologia , Psoríase/genética , Psoríase/patologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
4.
Cell Rep ; 31(11): 107771, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32553167

RESUMO

STING gain-of-function causes autoimmunity and immunodeficiency in mice and STING-associated vasculopathy with onset in infancy (SAVI) in humans. Here, we report that STING gain-of-function in mice prevents development of lymph nodes and Peyer's patches. We show that the absence of secondary lymphoid organs is associated with diminished numbers of innate lymphoid cells (ILCs), including lymphoid tissue inducer (LTi) cells. Although wild-type (WT) α4ß7+ progenitors differentiate efficiently into LTi cells, STING gain-of-function progenitors do not. Furthermore, STING gain-of-function impairs development of all types of ILCs. Patients with STING gain-of-function mutations have fewer ILCs, although they still have lymph nodes. In mice, expression of the STING mutant in RORγT-positive lineages prevents development of lymph nodes and reduces numbers of LTi cells. RORγT lineage-specific expression of STING gain-of-function also causes lung disease. Since RORγT is expressed exclusively in LTi cells during fetal development, our findings suggest that STING gain-of-function prevents lymph node organogenesis by reducing LTi cell numbers in mice.


Assuntos
Diferenciação Celular/imunologia , Imunidade Inata/imunologia , Linfonodos/imunologia , Linfócitos/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Mutação com Ganho de Função/imunologia , Tecido Linfoide/imunologia , Camundongos , Organogênese/imunologia
5.
Clin Immunol ; 216: 108466, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32470544

RESUMO

STING-associated vasculopathy with onset in infancy (SAVI) is an autoimmune disease caused by heterozygous gain of function mutations of STING (stimulator of interferon genes) that had initially been classified as a type I interferonopathy. We recently reported a genetically engineered mouse strain carrying a common SAVI-associated STING mutation. These STING N153S/WT mice reproduce key features of SAVI, including lung inflammation, loss of T cells in spleen and blood, splenomegaly and thymic hypoplasia. Here we show that αß T lymphocytopenia is due to disrupted T cell development and is associated with impaired T cell activation and a relative increase in γδ T cell numbers. These alterations were not rescued by additional knockout of the type I IFN receptor (IFNAR1). Collectively, our findings consolidate the concept that constitutive STING signalling leads to a SCID-like phenotype in STING N153S/WT mice.


Assuntos
Interferon Tipo I/imunologia , Proteínas de Membrana/imunologia , Doenças Vasculares/imunologia , Animais , Modelos Animais de Doenças , Feminino , Mutação com Ganho de Função/imunologia , Humanos , Inflamação/imunologia , Linfócitos Intraepiteliais/imunologia , Linfopenia/imunologia , Masculino , Camundongos , Camundongos SCID , Receptor de Interferon alfa e beta/imunologia
6.
J Clin Immunol ; 40(1): 82-95, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31686315

RESUMO

PURPOSE: Signal transducer and activator of transcription 1 (STAT1) is a transcription factor that mediates cellular responses to interferons (IFNs) and other cytokines and growth factors in diverse cell types. STAT1 gain-of-function (GOF) mutations result in an unexpectedly wide range of clinical features. It remains unclear why STAT1 GOF mutations result in such a broad spectrum of phenotypes. METHODS: We analyzed the clinical, molecular, and phenotypic characteristics of nine Chinese patients with STAT1 GOF mutations. RESULTS: This study enrolled nine patients with STAT1 GOF mutations including five novel mutations. We discuss the molecular and phenotypic characterization such as unique Penicillium marneffei lymphadenitis. Patients with STAT1 GOF mutations had defects in both innate and adaptive immunity, including impaired T cell receptor (TCR) diversity; reduced numbers of naïve and effector memory CD4+ T cells, memory B cells, and NK cells; and defects in the production of IL-17A and IFN-γ. In addition, experiments with primary immune cells revealed that enhanced STAT1 phosphorylation resulted from not only lower rates of STAT1 dephosphorylation but also increased total STAT1 expression. CONCLUSIONS: Our report provides the first comprehensive overview of the molecular genetics, clinical heterogeneity, and underlying immunological abnormalities of patients with STAT1 GOF mutations in China. In further study, to find the relationship between different STAT1 GOF mutations and clinical phenotype as well as the mechanism of increased total STAT1 expression will be needed.


Assuntos
Mutação com Ganho de Função/genética , Fator de Transcrição STAT1/genética , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , China , Feminino , Mutação com Ganho de Função/imunologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Células Matadoras Naturais/imunologia , Linfadenite/genética , Linfadenite/imunologia , Masculino , Penicillium/imunologia , Fenótipo , Fosforilação/genética , Fosforilação/imunologia , Fator de Transcrição STAT1/imunologia
7.
J Allergy Clin Immunol ; 141(5): 1844-1853.e2, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28859974

RESUMO

BACKGROUND: Sumoylation is a posttranslational reversible modification of cellular proteins through the conjugation of small ubiquitin-related modifier (SUMO) and comprises an important regulator of protein function. OBJECTIVE: We sought to characterize the molecular mechanism of a novel mutation at the SUMO motif on signal transducer and activator of transcription 1 (STAT1). METHODS: STAT1 sequencing and functional characterization were performed in transfection experiments by using immunoblotting and immunoprecipitation in STAT1-deficient cell lines. Transcriptional response and target gene activation were also investigated in PBMCs. RESULTS: We identified a novel STAT1 mutation (c.2114A>T, p.E705V) within the SUMO motif (702IKTE705) in a patient with disseminated Rhodococcus species infection, Norwegian scabies, chronic mucocutaneous candidiasis, hypothyroidism, and esophageal squamous cell carcinoma. The mutation is located in the tail segment and is predicted to disrupt STAT1 sumoylation. Immunoprecipitation experiments performed in transfected cells confirmed absent STAT1 sumoylation for E705V, whereas it was present in wild-type (WT) STAT1 cells, as well as the loss-of-function mutants L706S and Y701C. Furthermore, stimulation with IFN-γ led to enhanced STAT1 phosphorylation, enhanced transcriptional activity, and target gene expression in the E705V-transfected compared with WT-transfected cells. Computer modeling of WT and mutant STAT1 molecules showed variations in the accessibility of the phosphorylation site Y701, which corresponded to the loss-of-function and gain-of-function variants. CONCLUSION: This is the first report of a mutation in the STAT1 sumoylation motif associated with clinical disease. These data reinforce sumoylation as a key posttranslational regulatory modification of STAT1 and identify a novel mechanism for gain-of-function STAT1 disease in human subjects.


Assuntos
Mutação com Ganho de Função/imunologia , Mutação/genética , Fator de Transcrição STAT1/genética , Ubiquitina/genética , Animais , Células COS , Candidíase Mucocutânea Crônica/genética , Linhagem Celular , Chlorocebus aethiops , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Expressão Gênica/genética , Humanos , Fosforilação/genética , Proteína SUMO-1/genética , Sumoilação/genética , Transcrição Gênica/genética , Ativação Transcricional/genética , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA