Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Front Immunol ; 13: 870726, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35774795

RESUMO

Background: Inappropriate repair of DNA damage drives carcinogenesis. Lymphoid-specific helicase (HELLS) is an important component of the chromatin remodeling complex that helps repair DNA through various mechanisms such as DNA methylation, histone posttranslational modification, and nucleosome remodeling. Its role in human cancer initiation and progression has garnered recent attention. Our study aims to provide a more systematic and comprehensive understanding of the role of HELLS in the development and progression of multiple malignancies through analysis of HELLS in cancers. Methods: We explored the role of HELLS in cancers using The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) database. Multiple web platforms and software were used for data analysis, including R, Cytoscape, HPA, Archs4, TISIDB, cBioPortal, STRING, GSCALite, and CancerSEA. Results: High HELLS expression was found in a variety of cancers and differentially expressed across molecular and immune subtypes. HELLS was involved in many cancer pathways. Its expression positively correlated with Th2 and Tcm cells in most cancers. It also correlated with genetic markers of immunomodulators in various cancers. Conclusions: Our study elucidates the role HELLS plays in promotion, inhibition, and treatment of different cancers. HELLS is a potential cancer diagnostic and prognostic biomarker with immune, targeted, or cytotoxic therapeutic value. This work is a prerequisite to clinical validation and treatment of HELLS in cancers.


Assuntos
DNA Helicases , Neoplasias , Biologia Computacional , DNA Helicases/genética , DNA Helicases/imunologia , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/imunologia , Nucleossomos/genética , Prognóstico
2.
Oncologist ; 27(6): 501-511, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35278076

RESUMO

BACKGROUND: Thoracic SMARCA4-deficient undifferentiated tumors (SMARCA4-UT) are aggressive neoplasms. Data linking BAF alterations with tumor microenvironment (TME) and efficacy of immune checkpoint inhibitors (ICI) are contradictory. The TME of SMARCA4-UT and their response to ICI are unknown. MATERIALS AND METHODS: Patients diagnosed with SMARCA4-UT in our institution were included. Immunostainings for tertiary lymphoid structures (TLS), immune cell markers, and checkpoints were assessed. Validation was performed using an independent transcriptome dataset including SMARCA4-UT, non-small cell lung cancers (NSCLC) with/without SMARCA4 mutations, and unclassified thoracic sarcomas (UTS). CXCL9 and PD-L1 expressions were assessed in NSCLC and thoracic fibroblast cell lines, with/without SMARCA4 knockdown, treated with/without interferon gamma. RESULTS: Nine patients were identified. All samples but one showed no TLS, consistent with an immune desert TME phenotype. Four patients received ICI as part of their treatment, but the only one who responded, had a tumor with a TLS and immune-rich TME. Unsupervised clustering of the validation cohort using immune cell scores identified 2 clusters associated with cell ontogeny and immunity (cluster 1 enriched for NSCLC independently of SMARCA4 status (n = 9/10; P = .001); cluster 2 enriched for SMARCA4-UT (n = 11/12; P = .005) and UTS (n = 5/5; P = .0005). SMARCA4 loss-of-function experiments revealed interferon-induced upregulation of CXCL9 and PD-L1 expression in the NSCLC cell line with no effect on the thoracic fibroblast cell line. CONCLUSION: SMARCA4-UT mainly have an immune desert TME with limited efficacy to ICI. TME of SMARCA4-driven tumors varies according to the cell of origin questioning the interplay between BAF alterations, cell ontogeny and immunity.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , DNA Helicases , Inibidores de Checkpoint Imunológico , Neoplasias Pulmonares , Proteínas Nucleares , Sarcoma , Neoplasias de Tecidos Moles , Neoplasias Torácicas , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Biomarcadores Tumorais/imunologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , DNA Helicases/deficiência , DNA Helicases/imunologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Proteínas Nucleares/deficiência , Proteínas Nucleares/imunologia , Sarcoma/tratamento farmacológico , Sarcoma/imunologia , Sarcoma/patologia , Neoplasias de Tecidos Moles/tratamento farmacológico , Neoplasias de Tecidos Moles/imunologia , Neoplasias de Tecidos Moles/patologia , Neoplasias Torácicas/tratamento farmacológico , Neoplasias Torácicas/imunologia , Neoplasias Torácicas/patologia , Fatores de Transcrição/imunologia , Microambiente Tumoral/imunologia
3.
Fish Shellfish Immunol ; 121: 456-466, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35063603

RESUMO

The human zinc finger NFX1-type containing 1 (ZNFX1) is an interferon-stimulated protein associated to the outer mitochondrial membrane, able to bind dsRNAs and interact with MAVS proteins, promoting type I IFN response in the early stage of viral infection. An N-terminal Armadillo (ARM)-type fold and a large helicase core (P-loop) and zinc fingers confer RNA-binding and ATPase activities to ZNFX1. We studied the phylogenetic distribution of metazoan ZNFX1s, ZNFX1 gene expression trends and genomic and protein signatures during viral infection of invertebrates. Based on 221 ZNFX1 sequences, we obtained a polyphyletic tree with a taxonomy-consistent branching at the phylum-level only. In metazoan genomes, ZNFX1 genes were found either in single copy, with up to some tens of exons in vertebrates, or in multiple copies, with one or a few exons and one of them sometimes encompassing most of the coding sequence, in invertebrates like sponges, sea urchins and mollusks. Structural analyses of selected ZNFX1 proteins showed high conservation of the helicase region (P-loop), an overall conserved region and domain architecture, an ARM-fold mostly traceable, and the presence of intrinsically disordered regions of varying length and position. The remarkable over-expression of ZNFX1 in bivalve and gastropod mollusks infected with dsDNA viruses underscores the antiviral role of ZNFX1, whereas nothing similar was found in virus-infected nematodes and corals. Whether the functional diversification reported in the C. elegans ZNFX1 occurs in other metazoan proteins remains to be established.


Assuntos
DNA Helicases/imunologia , Imunidade Inata , Invertebrados , Viroses , Animais , Fatores de Restrição Antivirais/genética , Vírus de DNA/genética , Imunidade Inata/genética , Invertebrados/genética , Invertebrados/imunologia , Filogenia , Viroses/imunologia , Dedos de Zinco
4.
Signal Transduct Target Ther ; 7(1): 22, 2022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35075101

RESUMO

As a highly pathogenic human coronavirus, SARS-CoV-2 has to counteract an intricate network of antiviral host responses to establish infection and spread. The nucleic acid-induced stress response is an essential component of antiviral defense and is closely related to antiviral innate immunity. However, whether SARS-CoV-2 regulates the stress response pathway to achieve immune evasion remains elusive. In this study, SARS-CoV-2 NSP5 and N protein were found to attenuate antiviral stress granule (avSG) formation. Moreover, NSP5 and N suppressed IFN expression induced by infection of Sendai virus or transfection of a synthetic mimic of dsRNA, poly (I:C), inhibiting TBK1 and IRF3 phosphorylation, and restraining the nuclear translocalization of IRF3. Furthermore, HEK293T cells with ectopic expression of NSP5 or N protein were less resistant to vesicular stomatitis virus infection. Mechanistically, NSP5 suppressed avSG formation and disrupted RIG-I-MAVS complex to attenuate the RIG-I-mediated antiviral immunity. In contrast to the multiple targets of NSP5, the N protein specifically targeted cofactors upstream of RIG-I. The N protein interacted with G3BP1 to prevent avSG formation and to keep the cofactors G3BP1 and PACT from activating RIG-I. Additionally, the N protein also affected the recognition of dsRNA by RIG-I. This study revealed the intimate correlation between SARS-CoV-2, the stress response, and innate antiviral immunity, shedding light on the pathogenic mechanism of COVID-19.


Assuntos
Proteases 3C de Coronavírus/genética , Proteínas do Nucleocapsídeo de Coronavírus/genética , Proteína DEAD-box 58/genética , DNA Helicases/genética , Proteínas de Ligação a Poli-ADP-Ribose/genética , RNA Helicases/genética , Proteínas com Motivo de Reconhecimento de RNA/genética , Proteínas de Ligação a RNA/genética , Receptores Imunológicos/genética , SARS-CoV-2/genética , Grânulos de Estresse/genética , Animais , Chlorocebus aethiops , Proteases 3C de Coronavírus/imunologia , Proteínas do Nucleocapsídeo de Coronavírus/imunologia , Proteína DEAD-box 58/imunologia , DNA Helicases/imunologia , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Evasão da Resposta Imune , Fosfoproteínas/genética , Fosfoproteínas/imunologia , Poli I-C/farmacologia , Proteínas de Ligação a Poli-ADP-Ribose/imunologia , Ligação Proteica , RNA Helicases/imunologia , Proteínas com Motivo de Reconhecimento de RNA/imunologia , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/imunologia , Proteínas de Ligação a RNA/imunologia , Receptores Imunológicos/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Vírus Sendai/genética , Vírus Sendai/imunologia , Transdução de Sinais , Grânulos de Estresse/efeitos dos fármacos , Grânulos de Estresse/imunologia , Grânulos de Estresse/virologia , Células Vero , Vesiculovirus/genética , Vesiculovirus/imunologia
5.
J Clin Invest ; 132(2)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35040435

RESUMO

Inborn errors of nucleic acid metabolism often cause aberrant activation of nucleic acid sensing pathways, leading to autoimmune or autoinflammatory diseases. The SKIV2L RNA exosome is cytoplasmic RNA degradation machinery that was thought to be essential for preventing the self-RNA-mediated interferon (IFN) response. Here, we demonstrate the physiological function of SKIV2L in mammals. We found that Skiv2l deficiency in mice disrupted epidermal and T cell homeostasis in a cell-intrinsic manner independently of IFN. Skiv2l-deficient mice developed skin inflammation and hair abnormality, which were also observed in a SKIV2L-deficient patient. Epidermis-specific deletion of Skiv2l caused hyperproliferation of keratinocytes and disrupted epidermal stratification, leading to impaired skin barrier with no appreciable IFN activation. Moreover, Skiv2l-deficient T cells were chronically hyperactivated and these T cells attacked lesional skin as well as hair follicles. Mechanistically, SKIV2L loss activated the mTORC1 pathway in both keratinocytes and T cells. Both systemic and topical rapamycin treatment of Skiv2l-deficient mice ameliorated epidermal hyperplasia and skin inflammation. Together, we demonstrate that mTORC1, a classical nutrient sensor, also senses cytoplasmic RNA quality control failure and drives autoinflammatory disease. We also propose SKIV2L-associated trichohepatoenteric syndrome (THES) as a new mTORopathy for which sirolimus may be a promising therapy.


Assuntos
Doenças Autoimunes/imunologia , Citoplasma/imunologia , Diarreia Infantil/imunologia , Retardo do Crescimento Fetal/imunologia , Doenças do Cabelo/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/imunologia , Estabilidade de RNA/imunologia , RNA/imunologia , Animais , Doenças Autoimunes/genética , Citoplasma/genética , DNA Helicases/deficiência , DNA Helicases/imunologia , Diarreia Infantil/genética , Fácies , Retardo do Crescimento Fetal/genética , Doenças do Cabelo/genética , Inflamação/genética , Inflamação/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Camundongos Knockout , RNA/genética , Estabilidade de RNA/genética
6.
PLoS Pathog ; 18(1): e1010249, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35085371

RESUMO

Stress granules (SGs) are highly dynamic cytoplasmic foci that form in response to activation of the integrated stress response (ISR) that results in eIF2α phosphorylation and global translation shutdown. Stress granules, which are largely nucleated by G3BP1, serve as hubs for mRNA triage, but there is mounting evidence that they also perform cell signaling functions that are vital to cell survival, particularly during viral infection. We previously showed that SG formation leads to NFκB activation and JNK signaling and that this association may be due in part to G3BP1-dependent recruitment of PKR to SGs. Others have reported close associations between G3BP1 and various innate immune PRRs of the type 1 interferon signaling system, including RIG-I. We also reported SG assembly dynamics is dependent on the arginine-methylation status of G3BP1. Another protein that rapidly localizes to SGs, TDRD3, is a methyl reader protein that performs transcriptional activation and adaptor functions within the nucleus, but neither the mechanism nor its function in SGs is clear. Here, we present evidence that TDRD3 localizes to SGs partly based upon methylation potential of G3BP1. We also characterize granules that TDRD3 forms during overexpression and show that these granules can form in the absence of G3BP but also contain translation components found in canonical SGs. We also show for the first time that SGs recruit additional interferon effectors IRF3, IRF7, TBK1, and Sting, and provide evidence that TDRD3 may play a role in recruitment of these factors. We also present evidence that TDRD3 is a novel antiviral protein that is cleaved by enteroviral 2A proteinase. G3BP1 and TDRD3 knockdown in cells results in altered transcriptional regulation of numerous IFN effectors in complex modulatory patterns that are distinctive for G3BP1 and TDRD3. Overall, we describe a novel role of TDRD3 in innate immunity in which G3BP1 and TDRD3 may coordinate to play important roles in regulation of innate antiviral defenses.


Assuntos
DNA Helicases/imunologia , Imunidade Inata/imunologia , Proteínas de Ligação a Poli-ADP-Ribose/imunologia , Proteínas/imunologia , RNA Helicases/imunologia , Proteínas com Motivo de Reconhecimento de RNA/imunologia , Viroses/imunologia , Linhagem Celular , Humanos , Interferons/imunologia , Transdução de Sinais/imunologia , Grânulos de Estresse/imunologia
7.
Front Immunol ; 12: 762598, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34675941

RESUMO

Background: SMARCA4, the essential ATPase subunit of SWI/SNF chromatin remodeling complex, regulates transcription through the control of chromatin structure and is increasingly thought to play significant roles in human cancers. This study aims to explore the potential role of SMARCA4 with a view to providing insights on pathologic mechanisms implicated here. Methods: The potential roles of SMARCA4 in different tumors were explored based on The Cancer Genome Atlas (TCGA), Genotype-tissue expression (GTEx), Tumor Immune Estimation Resource (TIMER), and Gene Set Enrichment Analysis (GSEA) datasets. The expression difference, mutation and phosphorylation status, survival, pathological stage, DNA methylation, tumor mutation burden (TMB), microsatellite instability (MSI), mismatch repair (MMR), tumor microenvironment (TME), and immune cell infiltration related to SMARCA4 were analyzed. Results: High expression levels of SMARCA4 were observed in most cancer types. SMARCA4 expression in tumor samples correlates with poor overall survival in several cancers. Lung adenocarcinoma cases with altered SMARCA4 showed a poorer prognosis. Enhanced phosphorylation levels of S613, S695, S699, and S1417 were observed in several tumors, including breast cancer. SMARCA4 correlated with tumor immunity and associated with different immune cells and genes in different cancer types. TMB, MSI, MMR, and DNA methylation correlated with SMARCA4 dysregulation in cancers. SMARCA4 expression was negatively associated with CD8+ T-cell infiltration in several tumors. Furthermore, the SWI/SNF superfamily-type complex and ATPase complex may be involved in the functional mechanisms of SMARCA4, albeit these data require further confirmation. Conclusions: Our study offers a comprehensive understanding of the oncogenic roles of SMARCA4 across different tumors. SMARCA4 may correlate with tumor immunity.


Assuntos
DNA Helicases/genética , Neoplasias/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Linfócitos T CD8-Positivos/imunologia , DNA Helicases/imunologia , DNA Helicases/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Mutação , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/mortalidade , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Fosforilação , Prognóstico , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
8.
Front Immunol ; 12: 705848, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539636

RESUMO

Activation and differentiation of B cells depend on extensive rewiring of gene expression networks through changes in chromatin structure and accessibility. The chromatin remodeling complex BAF with its catalytic subunit Brg1 was previously identified as an essential regulator of early B cell development, however, how Brg1 orchestrates gene expression during mature B cell activation is less clear. Here, we find that Brg1 is required for B cell proliferation and germinal center formation through selective interactions with enhancers. Brg1 recruitment to enhancers following B cell activation was associated with increased chromatin accessibility and transcriptional activation of their coupled promoters, thereby regulating the expression of cell cycle-associated genes. Accordingly, Brg1-deficient B cells were unable to mount germinal center reactions and support the formation of class-switched plasma cells. Our findings show that changes in B cell transcriptomes that support B cell proliferation and GC formation depend on enhancer activation by Brg1. Thus, the BAF complex plays a critical role during the onset of the humoral immune response.


Assuntos
Linfócitos B/imunologia , Proliferação de Células , DNA Helicases , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica/imunologia , Centro Germinativo/imunologia , Proteínas Nucleares , Fatores de Transcrição , Animais , DNA Helicases/genética , DNA Helicases/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
9.
Int J Mol Sci ; 21(22)2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-33203071

RESUMO

Schimke immuno-osseous dysplasia (SIOD) is a rare multisystemic disorder with a variable clinical expressivity caused by biallelic variants in SMARCAL1. A phenotype-genotype correlation has been attempted and variable expressivity of biallelic SMARCAL1 variants may be associated with environmental and genetic disturbances of gene expression. We describe two siblings born from consanguineous parents with a diagnosis of SIOD revealed by whole exome sequencing (WES). Results: A homozygous missense variant in the SMARCAL1 gene (c.1682G>A; p.Arg561His) was identified in both patients. Despite carrying the same variant, the two patients showed substantial renal and immunological phenotypic differences. We describe features not previously associated with SIOD-both patients had congenital anomalies of the kidneys and of the urinary tract and one of them succumbed to a classical type congenital mesoblastic nephroma. We performed an extensive characterization of the immunophenotype showing combined immunodeficiency characterized by a profound lymphopenia, lack of thymic output, defective IL-7Rα expression, and disturbed B plasma cells differentiation and immunoglobulin production in addition to an altered NK-cell phenotype and function. Conclusions: Overall, our results contribute to extending the phenotypic spectrum of features associated with SMARCAL1 mutations and to better characterizing the underlying immunologic disorder with critical implications for therapeutic and management strategies.


Assuntos
Arteriosclerose , DNA Helicases , Rim , Células Matadoras Naturais/imunologia , Mutação de Sentido Incorreto , Nefroma Mesoblástico , Síndrome Nefrótica , Osteocondrodisplasias , Fenótipo , Doenças da Imunodeficiência Primária , Embolia Pulmonar , Sistema Urinário , Substituição de Aminoácidos , Arteriosclerose/diagnóstico por imagem , Arteriosclerose/genética , Arteriosclerose/imunologia , DNA Helicases/genética , DNA Helicases/imunologia , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-7/genética , Subunidade alfa de Receptor de Interleucina-7/imunologia , Rim/anormalidades , Rim/diagnóstico por imagem , Rim/imunologia , Masculino , Nefroma Mesoblástico/diagnóstico por imagem , Nefroma Mesoblástico/genética , Nefroma Mesoblástico/imunologia , Síndrome Nefrótica/diagnóstico por imagem , Síndrome Nefrótica/genética , Síndrome Nefrótica/imunologia , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/genética , Osteocondrodisplasias/imunologia , Doenças da Imunodeficiência Primária/diagnóstico por imagem , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/imunologia , Embolia Pulmonar/diagnóstico por imagem , Embolia Pulmonar/genética , Embolia Pulmonar/imunologia , Sistema Urinário/anormalidades , Sistema Urinário/diagnóstico por imagem , Sistema Urinário/imunologia , Sequenciamento Completo do Genoma
10.
Nat Immunol ; 21(8): 892-901, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32601470

RESUMO

Autoreactive T cells are eliminated in the thymus to prevent autoimmunity by promiscuous expression of tissue-restricted self-antigens in medullary thymic epithelial cells. This expression is dependent on the transcription factor Fezf2, as well as the transcriptional regulator Aire, but the entire picture of the transcriptional program has been obscure. Here, we found that the chromatin remodeler Chd4, also called Mi-2ß, plays a key role in the self-antigen expression in medullary thymic epithelial cells. To maximize the diversity of self-antigen expression, Fezf2 and Aire utilized completely distinct transcriptional mechanisms, both of which were under the control of Chd4. Chd4 organized the promoter regions of Fezf2-dependent genes, while contributing to the Aire-mediated induction of self-antigens via super-enhancers. Mice deficient in Chd4 specifically in thymic epithelial cells exhibited autoimmune phenotypes, including T cell infiltration. Thus, Chd4 plays a critical role in integrating Fezf2- and Aire-mediated gene induction to establish central immune tolerance.


Assuntos
Autoantígenos/imunologia , Tolerância Central/fisiologia , Regulação da Expressão Gênica/imunologia , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/imunologia , Animais , Autoantígenos/biossíntese , DNA Helicases/imunologia , DNA Helicases/metabolismo , Células HEK293 , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo , Proteína AIRE
11.
Nucleic Acids Res ; 48(1): 249-263, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31667508

RESUMO

Phage-inducible chromosomal island-like elements (PLEs) are bacteriophage satellites found in Vibrio cholerae. PLEs parasitize the lytic phage ICP1, excising from the bacterial chromosome, replicating, and mobilizing to new host cells following cell lysis. PLEs protect their host cell populations by completely restricting the production of ICP1 progeny. Previously, it was found that ICP1 replication was reduced during PLE(+) infection. Despite robust replication of the PLE genome, relatively few transducing units are produced. We investigated if PLE DNA replication itself is antagonistic to ICP1 replication. Here we identify key constituents of PLE replication and assess their role in interference of ICP1. PLE encodes a RepA_N initiation factor that is sufficient to drive replication from the PLE origin of replication during ICP1 infection. In contrast to previously characterized bacteriophage satellites, expression of the PLE initiation factor was not sufficient for PLE replication in the absence of phage. Replication of PLE was necessary for interference of ICP1 DNA replication, but replication of a minimalized PLE replicon was not sufficient for ICP1 DNA replication interference. Despite restoration of ICP1 DNA replication, non-replicating PLE remained broadly inhibitory against ICP1. These results suggest that PLE DNA replication is one of multiple mechanisms contributing to ICP1 restriction.


Assuntos
Bacteriófagos/genética , DNA Helicases/genética , DNA Bacteriano/genética , Vibrio cholerae/genética , Replicação Viral/genética , Bacteriófagos/metabolismo , Cromossomos Bacterianos/química , Cromossomos Bacterianos/imunologia , Cromossomos Bacterianos/virologia , DNA Helicases/imunologia , Replicação do DNA , DNA Bacteriano/imunologia , Lisogenia/genética , Origem de Replicação , Vibrio cholerae/imunologia , Vibrio cholerae/virologia
12.
Nat Commun ; 10(1): 3306, 2019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31341159

RESUMO

Memory CD8+ T cells have the ability to provide lifelong immunity against pathogens. Although memory features generally arise after challenge with a foreign antigen, naïve CD8 single positive (SP) thymocytes may acquire phenotypic and functional characteristics of memory cells in response to cytokines such as interleukin-4. This process is associated with the induction of the T-box transcription factor Eomesodermin (EOMES). However, the underlying molecular mechanisms remain ill-defined. Using epigenomic profiling, we show that these innate memory CD8SP cells acquire only a portion of the active enhancer repertoire of conventional memory cells. This reprograming is secondary to EOMES recruitment, mostly to RUNX3-bound enhancers. Furthermore, EOMES is found within chromatin-associated complexes containing BRG1 and promotes the recruitment of this chromatin remodelling factor. Also, the in vivo acquisition of EOMES-dependent program is BRG1-dependent. In conclusion, our results support a strong epigenetic basis for the EOMES-driven establishment of CD8+ T cell innate memory program.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Subunidade alfa 3 de Fator de Ligação ao Core/fisiologia , DNA Helicases/fisiologia , Epigênese Genética , Memória Imunológica , Proteínas Nucleares/fisiologia , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/fisiologia , Animais , Subunidade alfa 3 de Fator de Ligação ao Core/imunologia , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , DNA Helicases/imunologia , DNA Helicases/metabolismo , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Proteínas com Domínio T/genética , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
13.
mBio ; 10(3)2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31213553

RESUMO

The integrated stress response (ISR) is a cellular response system activated upon different types of stresses, including viral infection, to restore cellular homeostasis. However, many viruses manipulate this response for their own advantage. In this study, we investigated the association between murine norovirus (MNV) infection and the ISR and demonstrate that MNV regulates the ISR by activating and recruiting key ISR host factors. We observed that during MNV infection, there is a progressive increase in phosphorylated eukaryotic initiation factor 2α (p-eIF2α), resulting in the suppression of host translation, and yet MNV translation still progresses under these conditions. Interestingly, the shutoff of host translation also impacts the translation of key signaling cytokines such as beta interferon, interleukin-6, and tumor necrosis factor alpha. Our subsequent analyses revealed that the phosphorylation of eIF2α was mediated via protein kinase R (PKR), but further investigation revealed that PKR activation, phosphorylation of eIF2α, and translational arrest were uncoupled during infection. We further observed that stress granules (SGs) are not induced during MNV infection and that MNV can restrict SG nucleation and formation. We observed that MNV recruited the key SG nucleating protein G3BP1 to its replication sites and intriguingly the silencing of G3BP1 negatively impacts MNV replication. Thus, it appears that MNV utilizes G3BP1 to enhance replication but equally to prevent SG formation, suggesting an anti-MNV property of SGs. Overall, this study highlights MNV manipulation of SGs, PKR, and translational control to regulate cytokine translation and to promote viral replication.IMPORTANCE Viruses hijack host machinery and regulate cellular homeostasis to actively replicate their genome, propagate, and cause disease. In retaliation, cells possess various defense mechanisms to detect, destroy, and clear infecting viruses, as well as signal to neighboring cells to inform them of the imminent threat. In this study, we demonstrate that the murine norovirus (MNV) infection stalls host protein translation and the production of antiviral and proinflammatory cytokines. However, virus replication and protein translation still ensue. We show that MNV further prevents the formation of cytoplasmic RNA granules, called stress granules (SGs), by recruiting the key host protein G3BP1 to the MNV replication complex, a recruitment that is crucial to establishing and maintaining virus replication. Thus, MNV promotes immune evasion of the virus by altering protein translation. Together, this evasion strategy delays innate immune responses to MNV infection and accelerates disease onset.


Assuntos
Infecções por Caliciviridae/imunologia , Grânulos Citoplasmáticos/virologia , DNA Helicases/imunologia , Fator de Iniciação 2 em Eucariotos/imunologia , Evasão da Resposta Imune , Proteínas de Ligação a Poli-ADP-Ribose/imunologia , RNA Helicases/imunologia , Proteínas com Motivo de Reconhecimento de RNA/imunologia , eIF-2 Quinase/imunologia , Animais , Grânulos Citoplasmáticos/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Camundongos , Fosforilação , Biossíntese de Proteínas , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
14.
J Virol Methods ; 264: 11-17, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30381240

RESUMO

Hepatitis C Virus c33, a recombinant protein comprising residues 1192-1457 of NS3 helicase, has been a mainstay of HCV serology for decades. With seven unpaired cysteines, seroreactivity of E. coli expressed c33 is dependant on reductants. While engineering a c33 replacement for new anti-HCV serological tests, we sought to reduce oxidation sensitivity, a liability for immunodiagnostic reagent stability. A series of cysteine-to-serine substituted variants of a c33-like antigen was constructed and evaluated for reactivity against a panel of HCV-positive sera. Several variants were essentially nonreactive while others exhibited reactivity similar to or better than the wild-type construct. One demonstrated equivalent potency to wild-type but also diminished DTT dependence. To explore enhanced anti-NS3 reactivity, we constructed and examined an expanded series of antigens comprising individual helicase domains, the full-length helicase, additional cysteine-to-serine variants, and variants at positions critical to catalytic activity. Immunoassays using these latter NS3 helicase recombinants demonstrated that domain 1 possessed significantly more seroreactivity than previously believed, that the use of soluble full-length helicase protein enhanced sensitivity by several-fold over c33, and that anti-NS3 helicase seroreactivity was further enhanced by the introduction of point mutations which altered the catalytic activity or oxidation sensitivity of the antigen.


Assuntos
DNA Helicases/genética , DNA Helicases/imunologia , Hepacivirus/enzimologia , Hepacivirus/genética , Testes Sorológicos , Proteínas não Estruturais Virais/genética , Anticorpos Antivirais/sangue , Cisteína/genética , Cisteína/imunologia , DNA Helicases/metabolismo , Escherichia coli/genética , Engenharia Genética , Hepacivirus/imunologia , Humanos , Testes Imunológicos , Mutação Puntual , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Soroconversão , Proteínas não Estruturais Virais/imunologia
15.
Biochem Biophys Res Commun ; 503(4): 3212-3218, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30149919

RESUMO

The PI3K/Akt/mTOR pathway is thought to be closely associated with airway inflammation and is regulated by various upstream proteins. Brahma-related gene 1 (Brg1) plays an important role in chromatin remodeling and facilitates recruitment of essential transcription factors, leading to regulation of gene expression. Thus, the present study aimed at evaluating the anti-inflammatory role of Brg1 on house dust mite (HDM)-induced asthma through regulating the PI3K/Akt/mTOR pathway. The Brgfl/fl mice were crossbred with the SFTPC-Cre mice to generate bronchial epithelial cell specific Brg1 knockout mice, and LY294002 was used to inhibit PI3K. Western blot, immunofluorescence, immunoprecipitation, and immunohistochemical staining were used to detect the expression of proteins. An increase in Brg1 and a decrease in the PI3K/Akt/mTOR pathway activity were detected in asthmatic mice, but not in control mice. When Brg1 was knocked out, the asthma severity was ameliorated and the PI3K/Akt/mTOR pathway was activated. However, this protective effect could be suppressed by LY294002. Additionally, we observed that Brg1 was co-localized and co-immunoprecipitated with PI3K, using immunofluorescence and immunoprecipitation assays. Our results suggest that Brg1 might play an essential role in maintaining airway inflammation and affect the PI3K/Akt/mTOR pathway in asthma.


Assuntos
Asma/imunologia , DNA Helicases/imunologia , Proteínas Nucleares/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Transdução de Sinais , Serina-Treonina Quinases TOR/imunologia , Fatores de Transcrição/imunologia , Animais , Asma/patologia , Feminino , Inflamação/imunologia , Inflamação/patologia , Camundongos Endogâmicos C57BL
16.
Int J Cardiol ; 269: 250-258, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-30049497

RESUMO

BACKGROUND: Increased neutrophil infiltration and the ensuing inflammatory response represent a hallmark event in cardiac ischemia-reperfusion injury (IRI). It remains poorly defined how the epigenetic machinery contributes to this process. METHODS AND RESULTS: Here we report that mice with endothelial specific deletion of brahma related gene 1 (BRG1), a chromatin remodeling protein, exhibited amelioration when subjected to cardiac ischemia-reperfusion as evidenced by a reduction in infarct size as well as better recovery of heart function. Endothelial BRG1 deficiency also attenuated cardiac fibrosis following IRI when compared to wild type littermates. Interestingly, ablation of BRG1 in the endothelium suppressed neutrophil infiltration and down-regulated the levels of pro-inflammatory mediators in the heart following IRI. Further studies revealed that BRG1 activated the transcription of PODOCALYXIN (PODXL), an L-SELECTIN ligand crucial for neutrophil adhesion, in vascular endothelial cells in response to hypoxia-reoxygenation (HR). BRG1 knockdown by small interfering RNA abrogated HR-induced PODXL expression and blocked the adhesion of neutrophils to endothelial cells. Mechanistically, BRG1 alters the chromatin structure surrounding the PODXL promoter by interacting with JMJD2B, a histone H3K9 demethylase. Depletion of JMJD2B abrogated PODXL induction by HR and inhibited the adhesion of neutrophils to endothelial cells. CONCLUSION: Our data suggest that trans-activation of PODXL by the BRG1-JMJD2B complex in endothelial cells may promote neutrophil infiltration and consequently the pathogenesis of cardiac ischemia-reperfusion injury.


Assuntos
DNA Helicases/deficiência , Células Endoteliais/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Infiltração de Neutrófilos/fisiologia , Proteínas Nucleares/deficiência , Fatores de Transcrição/deficiência , Animais , Linhagem Celular Transformada , DNA Helicases/imunologia , Células Endoteliais/imunologia , Humanos , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/imunologia , Proteínas Nucleares/imunologia , Fatores de Transcrição/imunologia
17.
J Clin Invest ; 128(5): 2025-2041, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29664021

RESUMO

Cerebral white matter injury (WMI) persistently disrupts myelin regeneration by oligodendrocyte progenitor cells (OPCs). We identified a specific bioactive hyaluronan fragment (bHAf) that downregulates myelin gene expression and chronically blocks OPC maturation and myelination via a tolerance-like mechanism that dysregulates pro-myelination signaling via AKT. Desensitization of AKT occurs via TLR4 but not TLR2 or CD44. OPC differentiation was selectively blocked by bHAf in a maturation-dependent fashion at the late OPC (preOL) stage by a noncanonical TLR4/TRIF pathway that induced persistent activation of the FoxO3 transcription factor downstream of AKT. Activated FoxO3 selectively localized to oligodendrocyte lineage cells in white matter lesions from human preterm neonates and adults with multiple sclerosis. FoxO3 constraint of OPC maturation was bHAf dependent, and involved interactions at the FoxO3 and MBP promoters with the chromatin remodeling factor Brg1 and the transcription factor Olig2, which regulate OPC differentiation. WMI has adapted an immune tolerance-like mechanism whereby persistent engagement of TLR4 by bHAf promotes an OPC niche at the expense of myelination by engaging a FoxO3 signaling pathway that chronically constrains OPC differentiation.


Assuntos
Proteína Forkhead Box O3/imunologia , Tolerância Imunológica , Células Precursoras de Oligodendrócitos/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Animais , DNA Helicases/genética , DNA Helicases/imunologia , Feminino , Proteína Forkhead Box O3/genética , Humanos , Camundongos , Camundongos Knockout , Neuregulina-1/genética , Neuregulina-1/imunologia , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Células Precursoras de Oligodendrócitos/patologia , Fator de Transcrição 2 de Oligodendrócitos/genética , Fator de Transcrição 2 de Oligodendrócitos/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Receptores Toll-Like/genética , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
18.
Nat Immunol ; 19(2): 162-172, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29335648

RESUMO

Aire mediates the expression of tissue-specific antigens in thymic epithelial cells to promote tolerance against self-reactive T lymphocytes. However, the mechanism that allows expression of tissue-specific genes at levels that prevent harm is unknown. Here we show that Brg1 generates accessibility at tissue-specific loci to impose central tolerance. We found that Aire has an intrinsic repressive function that restricts chromatin accessibility and opposes Brg1 across the genome. Aire exerted this repressive influence within minutes after recruitment to chromatin and restrained the amplitude of active transcription. Disease-causing mutations that impair Aire-induced activation also impair the protein's repressive function, which indicates dual roles for Aire. Together, Brg1 and Aire fine-tune the expression of tissue-specific genes at levels that prevent toxicity yet promote immune tolerance.


Assuntos
Tolerância Central/imunologia , DNA Helicases/imunologia , Regulação da Expressão Gênica/imunologia , Proteínas Nucleares/imunologia , Timo/imunologia , Fatores de Transcrição/imunologia , Animais , Cromatina , Camundongos , Camundongos Transgênicos , Proteína AIRE
19.
Rheumatology (Oxford) ; 57(4): 712-717, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29294089

RESUMO

Objectives: Autoantibodies targeting ubiquitously expressed nuclear antigens can be identified in most patients with SSc. Cytoplasmic autoantibodies (in otherwise ANA-negative sera) targeting eukaryotic initiation factor-2B (anti-eIF2B) have recently been identified in SSc with clinical associations to dcSSc disease and interstitial lung disease (ILD), although the majority of samples originated from a tertiary SSc-ILD centre. We investigated the prevalence and clinical associations of recently described SSc-specific (including anti-eIF2B) and other cytoplasmic autoantibodies in ANA-negative sera obtained from a large representative SSc cohort. Methods: ANA-negative sera from the Scleroderma Family Registry and DNA Repository underwent indirect immunofluorescence, radiolabelled protein immunoprecipitation (± immunodepletion) to identify anti-eIF2B and other CTD-related autoantibodies. The clinical phenotype of positive samples was evaluated. Results: Immunoprecipitation was performed on 128 ANA-negative samples (obtained from 3249 SSc patients). Anti-eIF2B antibodies were present in nine patients (7%), the majority of whom had dcSSc (8/9). SSc-ILD was present in all anti-eIF2B patients for whom chest imaging was available (7/9). Anti-synthetase autoantibodies (targeting PL12, PL7, OJ and Zo) were identified in seven patients (5.5%), all of whom fulfilled the 2013 ACR/EULAR classification criteria for SSc and had evidence of SSc-ILD where relevant outcomes were available for evaluation. Anti-RuvBL1/2 antibodies were identified in two patients with SSc-overlap syndromes. Conclusion: Anti-eIF2B antibodies are cytoplasmic SSc-specific autoantibodies with strong clinical associations with dcSSc and SSc-ILD found in ANA-negative sera. Anti-synthetase autoantibodies, and other recently discovered SSc-specific antibodies such as anti-RuvBL1/2, can also be identified in ANA-negative SSc.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/imunologia , Autoanticorpos/imunologia , Proteínas de Transporte/imunologia , DNA Helicases/imunologia , Fator de Iniciação 2B em Eucariotos/imunologia , Ligases/imunologia , Escleroderma Sistêmico/imunologia , ATPases Associadas a Diversas Atividades Celulares/sangue , Anticorpos Antinucleares/sangue , Anticorpos Antinucleares/imunologia , Autoanticorpos/sangue , Proteínas de Transporte/sangue , DNA Helicases/sangue , Fator de Iniciação 2B em Eucariotos/sangue , Humanos , Imunoprecipitação , Ligases/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA