Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Immunol ; 195(3): 1025-33, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26101320

RESUMO

Because of their unique capacity to cross-present Ags to CD8(+) T cells, mouse lymphoid tissue-resident CD8(+) dendritic cells (DCs) and their migratory counterparts are critical for priming antiviral T cell responses. High expression of the dsRNA sensor TLR3 is a distinctive feature of these cross-presenting DC subsets. TLR3 engagement in CD8(+) DCs promotes cross-presentation and the acquisition of effector functions required for driving antiviral T cell responses. In this study, we performed a comprehensive analysis of the TLR3-induced antiviral program and cell-autonomous immunity in CD8(+) DC lines and primary CD8(+) DCs. We found that TLR3-ligand polyinosinic-polycytidylic acid and human rhinovirus infection induced a potent antiviral protection against Sendai and vesicular stomatitis virus in a TLR3 and type I IFN receptor-dependent manner. Polyinosinic-polycytidylic acid-induced antiviral genes were identified by mass spectrometry-based proteomics and transcriptomics in the CD8(+) DC line. Nanostring nCounter experiments confirmed that these antiviral genes were induced by TLR3 engagement in primary CD8(+) DCs, and indicated that many are secondary TLR3-response genes requiring autocrine IFN-ß stimulation. TLR3-activation thus establishes a type I IFN-dependent antiviral program in a DC subtype playing crucial roles in priming adaptive antiviral immune responses. This mechanism is likely to shield the priming of antiviral responses against inhibition or abrogation by the viral infection. It could be particularly relevant for viruses detected mainly by TLR3, which may not trigger type I IFN production by DCs that lack TLR3, such as plasmacytoid DCs or CD8(-) DCs.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Interferon beta/imunologia , Receptor 3 Toll-Like/imunologia , Animais , Apresentação Cruzada/imunologia , Humanos , Interferon beta/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/virologia , Poli I-C/imunologia , Receptor de Interferon alfa e beta/imunologia , Rhinovirus/imunologia , Vírus Sendai/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia
2.
Mol Ther ; 24(9): 1675-85, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27377043

RESUMO

Cell penetrating peptides (CPPs) from the protein ZEBRA are promising candidates to exploit in therapeutic cancer vaccines, since they can transport antigenic cargos into dendritic cells and induce tumor-specific T cells. Employing CPPs for a given cancer indication will require engineering to include relevant tumor-associated epitopes, administration with an appropriate adjuvant, and testing for antitumor immunity. We assessed the importance of structural characteristics, efficiency of in vitro transduction of target cells, and choice of adjuvant in inducing the two key elements in antitumor immunity, CD4 and CD8 T cells, as well as control of tumor growth in vivo. Structural characteristics associated with CPP function varied according to CPP truncations and cargo epitope composition, and correlated with in vitro transduction efficiency. However, subsequent in vivo capacity to induce CD4 and CD8 T cells was not always predicted by in vitro results. We determined that the critical parameter for in vivo efficacy using aggressive mouse tumor models was the choice of adjuvant. Optimal pairing of a particular ZEBRA-CPP sequence and antigenic cargo together with adjuvant induced potent antitumor immunity. Our results highlight the irreplaceable role of in vivo testing of novel vaccine constructs together with adjuvants to select combinations for further development.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/imunologia , Peptídeos Penetradores de Células/imunologia , Neoplasias/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/química , Dicroísmo Circular , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Camundongos , Neoplasias/mortalidade , Neoplasias/patologia , Neoplasias/terapia , Transativadores/química , Transativadores/imunologia , Resultado do Tratamento , Vacinação
3.
J Immunol ; 193(4): 1690-700, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25009204

RESUMO

Arginine, a semiessential amino acid implicated in diverse cellular processes, is a substrate for two arginases-Arg1 and Arg2-having different expression patterns and functions. Although appropriately regulated Arg1 expression is critical for immune responses, this has not been documented for Arg2. We show that Arg2 is the dominant enzyme in dendritic cells (DCs) and is repressed by microRNA-155 (miR155) during their maturation. miR155 is known to be strongly induced in various mouse and human DC subsets in response to diverse maturation signals, and miR155-deficient DCs exhibit an impaired ability to induce Ag-specific T cell responses. By means of expression profiling studies, we identified Arg2 mRNA as a novel miR155 target in mouse DCs. Abnormally elevated levels of Arg2 expression and activity were observed in activated miR155-deficient DCs. Conversely, overexpression of miR155 inhibited Arg2 expression. Bioinformatic and functional analyses confirmed that Arg2 mRNA is a direct target of miR155. Finally, in vitro and in vivo functional assays using DCs exhibiting deregulated Arg2 expression indicated that Arg2-mediated arginine depletion in the extracellular milieu impairs T cell proliferation. These results indicate that miR155-induced repression of Arg2 expression is critical for the ability of DCs to drive T cell activation by controlling arginine availability in the extracellular environment.


Assuntos
Arginase/biossíntese , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/enzimologia , Ativação Linfocitária/imunologia , MicroRNAs/genética , Animais , Arginase/antagonistas & inibidores , Arginase/genética , Arginina/metabolismo , Proliferação de Células , Células Dendríticas/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Células HEK293 , Humanos , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Processamento Pós-Transcricional do RNA , RNA Mensageiro/genética
4.
J Immunol ; 193(6): 2743-52, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25114100

RESUMO

Autoimmune neuroinflammation, including multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), a prototype for T cell-mediated autoimmunity, is believed to result from immune tolerance dysfunction leading to demyelination and substantial neurodegeneration. We previously showed that CNS-restricted expression of hepatocyte growth factor (HGF), a potent neuroprotective factor, reduced CNS inflammation and clinical deficits associated with EAE. In this study, we demonstrate that systemic HGF treatment ameliorates EAE through the development of tolerogenic dendritic cells (DCs) with high expression levels of glucocorticoid-induced leucine zipper (GILZ), a transcriptional repressor of gene expression and a key endogenous regulator of the inflammatory response. RNA interference-directed neutralization of GILZ expression by DCs suppressed the induction of tolerance caused by HGF. Finally, adoptive transfer of HGF-treated DCs from wild-type but not GILZ gene-deficient mice potently mediated functional recovery in recipient mice with established EAE through effective modulation of autoaggressive T cell responses. Altogether, these results show that by inducing GILZ in DCs, HGF reproduces the mechanism of immune regulation induced by potent immunomodulatory factors such as IL-10, TGF-ß1, and glucocorticoids and therefore that HGF therapy may have potential in the treatment of autoimmune dysfunctions.


Assuntos
Corticosterona/imunologia , Encefalomielite Autoimune Experimental/imunologia , Fator de Crescimento de Hepatócito/farmacologia , Linfócitos T/imunologia , Fatores de Transcrição/biossíntese , Transferência Adotiva , Animais , Autoimunidade/imunologia , Proliferação de Células , Células Cultivadas , Sistema Nervoso Central/imunologia , Corticosterona/sangue , Células Dendríticas/imunologia , Células Dendríticas/transplante , Encefalomielite Autoimune Experimental/tratamento farmacológico , Feminino , Fator de Crescimento de Hepatócito/biossíntese , Tolerância Imunológica/genética , Inflamação/imunologia , Interleucina-10/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/imunologia , Interferência de RNA , RNA Interferente Pequeno , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta1/imunologia
5.
Nucleic Acids Res ; 42(15): 9641-55, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25104025

RESUMO

The activation, or maturation, of dendritic cells (DCs) is crucial for the initiation of adaptive T-cell mediated immune responses. Research on the molecular mechanisms implicated in DC maturation has focused primarily on inducible gene-expression events promoting the acquisition of new functions, such as cytokine production and enhanced T-cell-stimulatory capacity. In contrast, mechanisms that modulate DC function by inducing widespread gene-silencing remain poorly understood. Yet the termination of key functions is known to be critical for the function of activated DCs. Genome-wide analysis of activation-induced histone deacetylation, combined with genome-wide quantification of activation-induced silencing of nascent transcription, led us to identify a novel inducible transcriptional-repression pathway that makes major contributions to the DC-maturation process. This silencing response is a rapid primary event distinct from repression mechanisms known to operate at later stages of DC maturation. The repressed genes function in pivotal processes--including antigen-presentation, extracellular signal detection, intracellular signal transduction and lipid-mediator biosynthesis--underscoring the central contribution of the silencing mechanism to rapid reshaping of DC function. Interestingly, promoters of the repressed genes exhibit a surprisingly high frequency of PU.1-occupied sites, suggesting a novel role for this lineage-specific transcription factor in marking genes poised for inducible repression.


Assuntos
Células Dendríticas/metabolismo , Inativação Gênica , Proteínas Nucleares/genética , Transativadores/genética , Transcrição Gênica , Animais , Humanos , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo
6.
EMBO J ; 28(8): 1111-20, 2009 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-19279666

RESUMO

Nucleotide excision repair (NER) requires the coordinated sequential assembly and actions of the involved proteins at sites of DNA damage. Following damage recognition, dual incision 5' to the lesion by ERCC1-XPF and 3' to the lesion by XPG leads to the removal of a lesion-containing oligonucleotide of about 30 nucleotides. The resulting single-stranded DNA (ssDNA) gap on the undamaged strand is filled in by DNA repair synthesis. Here, we have asked how dual incision and repair synthesis are coordinated in human cells to avoid the exposure of potentially harmful ssDNA intermediates. Using catalytically inactive mutants of ERCC1-XPF and XPG, we show that the 5' incision by ERCC1-XPF precedes the 3' incision by XPG and that the initiation of repair synthesis does not require the catalytic activity of XPG. We propose that a defined order of dual incision and repair synthesis exists in human cells in the form of a 'cut-patch-cut-patch' mechanism. This mechanism may aid the smooth progression through the NER pathway and contribute to genome integrity.


Assuntos
Dano ao DNA , Reparo do DNA , DNA/metabolismo , Animais , Domínio Catalítico , Linhagem Celular , DNA/genética , DNA/efeitos da radiação , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endonucleases/genética , Endonucleases/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Raios Ultravioleta
7.
Blood ; 118(7): 1838-44, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21642598

RESUMO

The bone marrow (BM) is an organ extremely efficient in mediating long-term survival of plasma cells (PCs), ensuring an immune humoral memory. This implies that the BM must provide continuously key PC survival factors. Our results show that the BM is an organ constitutively rich in a proliferation-inducing ligand (APRIL), a member of the tumor necrosis factor superfamily implicated in PC survival. APRIL production is induced during hematopoiesis in myeloid cells by non-lineage-committing factors such as stem cell factor, thrombopoietin, IL-3, and FMS-like tyrosine kinase 3 ligand. Notably, APRIL production, both in the human and mouse systems, peaks in myeloid precursor cells, before dropping in fully mature granulocytes. Myeloid cells secrete APRIL that circulates freely in BM plasma to act on PCs, usually at distance from APRIL production sites. Selective APRIL in vivo antagonism and in vitro coculture experiments further demonstrated that myeloid precursor cells mediates PC survival in an APRIL-dependent manner Thus, APRIL production by myeloid precursor cells shows that the 2 main BM functions, hematopoiesis and long-term PC survival, are linked. Such constitutive and high APRIL production may explain why BM mediates long-term PC survival.


Assuntos
Células da Medula Óssea/citologia , Leucopoese , Células Mieloides/citologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Células da Medula Óssea/metabolismo , Sobrevivência Celular , Células Cultivadas , Técnicas de Inativação de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Plasmócitos/citologia , Plasmócitos/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
8.
Blood ; 117(17): 4490-500, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21385848

RESUMO

MicroRNAs (miRNAs) are small, noncoding RNAs that regulate target mRNAs by binding to their 3' untranslated regions. There is growing evidence that microRNA-155 (miR155) modulates gene expression in various cell types of the immune system and is a prominent player in the regulation of innate and adaptive immune responses. To define the role of miR155 in dendritic cells (DCs) we performed a detailed analysis of its expression and function in human and mouse DCs. A strong increase in miR155 expression was found to be a general and evolutionarily conserved feature associated with the activation of DCs by diverse maturation stimuli in all DC subtypes tested. Analysis of miR155-deficient DCs demonstrated that miR155 induction is required for efficient DC maturation and is critical for the ability of DCs to promote antigen-specific T-cell activation. Expression-profiling studies performed with miR155(-/-) DCs and DCs overexpressing miR155, combined with functional assays, revealed that the mRNA encoding the transcription factor c-Fos is a direct target of miR155. Finally, all of the phenotypic and functional defects exhibited by miR155(-/-) DCs could be reproduced by deregulated c-Fos expression. These results indicate that silencing of c-Fos expression by miR155 is a conserved process that is required for DC maturation and function.


Assuntos
Células Dendríticas/fisiologia , Inativação Gênica/imunologia , MicroRNAs/imunologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem Celular , Células Dendríticas/citologia , Evolução Molecular , Humanos , Camundongos , Camundongos Mutantes , MicroRNAs/genética , Monócitos/citologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia
9.
Proc Natl Acad Sci U S A ; 106(8): 2735-40, 2009 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19193853

RESUMO

In response to inflammatory stimulation, dendritic cells (DCs) have a remarkable pattern of differentiation (maturation) that exhibits specific mechanisms to control immunity. Here, we show that in response to Lipopolysaccharides (LPS), several microRNAs (miRNAs) are regulated in human monocyte-derived dendritic cells. Among these miRNAs, miR-155 is highly up-regulated during maturation. Using LNA silencing combined to microarray technology, we have identified the Toll-like receptor/interleukin-1 (TLR/IL-1) inflammatory pathway as a general target of miR-155. We further demonstrate that miR-155 directly controls the level of TAB2, an important signal transduction molecule. Our observations suggest, therefore, that in mature human DCs, miR-155 is part of a negative feedback loop, which down-modulates inflammatory cytokine production in response to microbial stimuli.


Assuntos
Células Dendríticas/metabolismo , Interleucina-1/metabolismo , MicroRNAs/fisiologia , Monócitos/metabolismo , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Bases , Células Cultivadas , Primers do DNA , Humanos , Ativação Linfocitária , Processamento Pós-Transcricional do RNA , Receptores Toll-Like/metabolismo
10.
J Immunol ; 182(12): 8094-103, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494335

RESUMO

Endogenous retroviruses are implicated in the pathogenesis of systemic lupus erythematosus (SLE). Because four different classes of endogenous retroviruses, i.e., ecotropic, xenotropic, polytropic, or modified polytropic (mPT), are expressed in mice, we investigated the possibility that a particular class of endogenous retroviruses is associated with the development of murine SLE. We observed >15-fold increased expression of mPT env (envelope) RNA in livers of all four lupus-prone mice, as compared with those of nine nonautoimmune strains of mice. This was not the case for the three other classes of retroviruses. Furthermore, we found that in addition to intact mPT transcripts, many strains of mice expressed two defective mPT env transcripts which carry a deletion in the env sequence of the 3' portion of the gp70 surface protein and the 5' portion of the p15E transmembrane protein, respectively. Remarkably, in contrast to nonautoimmune strains of mice, all four lupus-prone mice expressed abundant levels of intact mPT env transcripts, but only low or nondetectable levels of the mutant env transcripts. The Sgp3 (serum gp70 production 3) locus derived from lupus-prone mice was responsible for the selective up-regulation of the intact mPT env RNA. Finally, we observed that single-stranded RNA-specific TLR7 played a critical role in the production of anti-gp70 autoantibodies. These data suggest that lupus-prone mice may possess a unique genetic mechanism responsible for the expression of mPT retroviruses, which could act as a triggering factor through activating TLR7 for the development of autoimmune responses in mice predisposed to SLE.


Assuntos
Retrovirus Endógenos/metabolismo , Produtos do Gene env/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , RNA Viral/genética , Regulação para Cima , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , Retrovirus Endógenos/genética , Produtos do Gene env/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação/genética , Alinhamento de Sequência , Receptor 7 Toll-Like/metabolismo , Transcrição Gênica/genética
11.
Nucleic Acids Res ; 37(8): 2514-28, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19264803

RESUMO

Nucleosome depletion at transcription start sites (TSS) has been documented genome-wide in multiple eukaryotic organisms. However, the mechanisms that mediate this nucleosome depletion and its functional impact on transcription remain largely unknown. We have studied these issues at human MHC class II (MHCII) genes. Activation-induced nucleosome free regions (NFR) encompassing the TSS were observed at all MHCII genes. Nucleosome depletion was exceptionally strong, attaining over 250-fold, at the promoter of the prototypical HLA-DRA gene. The NFR was induced primarily by the transcription factor complex that assembles on the conserved promoter-proximal enhancer situated upstream of the TSS. Functional analyses performed in the context of native chromatin demonstrated that displacing the NFR without altering the sequence of the core promoter induced a shift in the position of the TSS. The NFR thus appears to play a critical role in transcription initiation because it directs correct TSS positioning in vivo. Our results provide support for a novel mechanism in transcription initiation whereby the position of the TSS is controlled by nucleosome eviction rather than by promoter sequence.


Assuntos
Genes MHC da Classe II , Nucleossomos/metabolismo , Regiões Promotoras Genéticas , Sítio de Iniciação de Transcrição , Linfócitos B/imunologia , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina , Antígenos HLA-DR/genética , Cadeias alfa de HLA-DR , Humanos , Interferon gama/farmacologia
12.
J Autoimmun ; 35(2): 153-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20619604

RESUMO

The endogenous retroviral envelope glycoprotein, gp70, implicated in murine systemic lupus erythematosus (SLE), has been considered to be a product of xenotropic, polytropic (PT) and modified PT (mPT) endogenous retroviruses. It is secreted by hepatocytes like an acute phase protein, but its response is under a genetic control. Given critical roles of TLR7 and TLR9 in the pathogenesis of SLE, we assessed their contribution to the acute phase expression of serum gp70, and defined a pivotal role of the Sgp3 (serum gp70 production 3) and Sgp4 loci in this response. Our results demonstrated that serum levels of gp70 were up-regulated in lupus-prone NZB mice injected with TLR7 or TLR9 agonist at levels comparable to those induced by injection of IL-1, IL-6 or TNF. In addition, studies of C57BL/6 Sgp3 and/or Sgp4 congenic mice defined the major roles of these two loci in up-regulated production of serum gp70 during acute phase responses. Finally, the analysis of Sgp3 congenic mice strongly suggests the presence of at least two distinct genetic factors in the Sgp3 interval, one of which controlled the basal-level expression of xenotropic, PT and mPT gp70 and the other which controlled the up-regulated production of xenotropic and mPT gp70 during acute phase responses. Our results uncovered an additional pathogenic role of TLR7 and TLR9 in murine lupus nephritis by promoting the expression of nephritogenic gp70 autoantigen. Furthermore, they revealed the involvement of multiple regulatory genes for the expression of gp70 autoantigen under steady-state and inflammatory conditions in lupus-prone mice.


Assuntos
Proteínas de Fase Aguda/biossíntese , Reação de Fase Aguda/genética , Autoantígenos/biossíntese , Retrovirus Endógenos/genética , Glicoproteínas/biossíntese , Glicoproteínas/metabolismo , Lúpus Eritematoso Sistêmico/genética , Chaperonas Moleculares/metabolismo , Proteínas Virais/biossíntese , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/imunologia , Reação de Fase Aguda/sangue , Reação de Fase Aguda/complicações , Reação de Fase Aguda/imunologia , Reação de Fase Aguda/virologia , Animais , Autoantígenos/sangue , Autoantígenos/genética , Autoantígenos/imunologia , Citocinas/administração & dosagem , Modelos Animais de Doenças , Feminino , Regulação Viral da Expressão Gênica , Glicoproteínas/sangue , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Lipopolissacarídeos/administração & dosagem , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/virologia , Nefrite Lúpica/etiologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Transgênicos , Chaperonas Moleculares/genética , Oligodesoxirribonucleotídeos/administração & dosagem , Receptor 7 Toll-Like/imunologia , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/imunologia , Receptor Toll-Like 9/metabolismo , Proteínas Virais/sangue , Proteínas Virais/genética , Proteínas Virais/imunologia
13.
J Autoimmun ; 34(4): 339-48, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19944565

RESUMO

Accumulating evidence supports the idea that TLR7 and TLR9 play pathogenic and protective roles, respectively, in the development of murine systemic lupus erythematosus (SLE). However, the molecular mechanism responsible for the accelerated development of SLE resulting from the deletion of TLR9 and the respective contributions of TLR7 and TLR9 to the development of different autoimmune responses against nuclear and non-nuclear autoantigens implicated in lupus nephritis have not been well defined. In the present study, we addressed these questions by assessing the effect of the TLR9 and/or TLR7 deletion on the production of various autoantibodies and the development of lupus nephritis in C57BL/6 mice congenic for the Nba2 (NZB autoimmunity 2) locus (B6.Nba2). TLR9-deficient B6.Nba2 mice displayed increased production of autoantibodies against nuclear antigens, serum retroviral gp70 and glomerular matrix antigens, and developed a markedly accelerated form of lupus nephritis. Enhanced disease was associated with functionally upregulated expression of TLR7, as documented by an increased TLR7-dependent activation of B cells and plasmacytoid dendritic cells. Notably, disease exacerbation in TLR9-deficient mice was completely suppressed by the deletion of TLR7. Our results indicate that TLR7 has a pivotal role in a wide variety of autoimmune responses against DNA- and RNA-containing nuclear antigens, retroviral gp70 and glomerular matrix antigens implicated in murine SLE, and that enhanced TLR7 activity is critical for the accelerated development of SLE in TLR9-deficient lupus-prone mice.


Assuntos
Lúpus Eritematoso Sistêmico/etiologia , Glicoproteínas de Membrana/fisiologia , Receptor 7 Toll-Like/fisiologia , Receptor Toll-Like 9/deficiência , Animais , Antígenos Nucleares/imunologia , Autoanticorpos/biossíntese , Autoantígenos/imunologia , Autoimunidade , Progressão da Doença , Membrana Basal Glomerular/imunologia , Glomérulos Renais/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Nefrite Lúpica/etiologia , Camundongos , Camundongos Knockout
14.
Nucleic Acids Res ; 35(2): 595-605, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17175541

RESUMO

The small GTPase RAB4 regulates endocytic recycling, a process that contributes to Major Histocompatibility Complex (MHC)-mediated antigen presentation by specialized antigen presenting cells (APC) of the immune system. The gene encoding the RAB4B isoform of RAB4 was singled out by two complementary genome-wide screens. One of these consisted of a computer scan to identify genes containing characteristic MHC class II-related regulatory sequences. The second was the use of chromatin immunoprecipitation coupled to microarrays (ChIP-on-chip) to identify novel targets of a transcriptional co-activator called the MHC class II transactivator (CIITA). We show that the RAB4B gene is regulated by a typical MHC class II-like enhancer that is controlled directly by both CIITA and the multiprotein transcription factor complex known as the MHC class II enhanceosome. RAB4B expression is thus activated by the same regulatory machinery that is known to be essential for the expression of MHC class II genes. This molecular link between the transcriptional activation of RAB4B and MHC class II genes implies that APC boost their antigen presentation capacity by increasing RAB4-mediated endocytic recycling.


Assuntos
Elementos Facilitadores Genéticos , Genes MHC da Classe II , Ativação Transcricional , Proteínas rab4 de Ligação ao GTP/genética , Sítios de Ligação , Células Cultivadas , Proteínas de Ligação a DNA , Células Dendríticas/imunologia , Endocitose , Genômica , Humanos , Interferon gama/farmacologia , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição de Fator Regulador X , Transativadores/metabolismo , Fatores de Transcrição , Proteínas rab4 de Ligação ao GTP/biossíntese
15.
Nucleic Acids Res ; 35(10): 3431-41, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17478518

RESUMO

Posttranslational histone modifications associated with actively expressed genes are generally believed to be introduced primarily by histone-modifying enzymes that are recruited by transcription factors or their associated co-activators. We have performed a comprehensive spatial and temporal analyses of the histone modifications that are deposited upon activation of the MHC class II gene HLA-DRA by the co-activator CIITA. We find that transcription-associated histone modifications are introduced during two sequential phases. The first phase precedes transcription initiation and is characterized exclusively by a rapid increase in histone H4 acetylation over a large upstream domain. All other modifications examined, including the acetylation and methylation of several residues in histone H3, are restricted to short regions situated at or within the 5' end of the gene and are established during a second phase that is concomitant with ongoing transcription. This second phase is completely abrogated when elongation by RNA polymerase II is blocked. These results provide strong evidence that transcription elongation can play a decisive role in the deposition of histone modification patterns associated with inducible gene activation.


Assuntos
Genes MHC da Classe II , Antígenos HLA-DR/genética , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Ativação Transcricional , Região 5'-Flanqueadora , Acetilação , Linhagem Celular , Antígenos HLA-DR/biossíntese , Cadeias alfa de HLA-DR , Humanos , Interferon gama/farmacologia , Cinética , Metilação
16.
Nucleic Acids Res ; 35(9): 3053-63, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17452369

RESUMO

FEN-1 and XPG are members of the FEN-1 family of structure-specific nucleases, which share a conserved active site. FEN-1 plays a central role in DNA replication, whereas XPG is involved in nucleotide excision repair (NER). Both FEN-1 and XPG are active on flap structures, but only XPG cleaves bubble substrates. The spacer region of XPG is dispensable for nuclease activity on flap substrates but is required for NER activity and for efficient processing of bubble substrates. Here, we inserted the spacer region of XPG between the nuclease domains of FEN-1 to test whether this domain would be sufficient to confer XPG-like substrate specificity and NER activity on a related nuclease. The resulting FEN-1-XPG hybrid protein is active on flap and, albeit at low levels, on bubble substrates. Like FEN-1, the activity of FEN-1-XPG was stimulated by a double-flap substrate containing a 1-nt 3' flap, whereas XPG does not show this substrate preference. Although no NER activity was detected in vitro, the FEN-1-XPG hybrid displays substantial NER activity in vivo. Hence, insertion of the XPG spacer region into FEN-1 results in a hybrid protein with biochemical properties reminiscent of both nucleases, including partial NER activity.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Endonucleases/química , Endonucleases/metabolismo , Endonucleases Flap/química , Endonucleases Flap/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Células Cultivadas , Dano ao DNA , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Endonucleases Flap/genética , Humanos , Proteínas Nucleares/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato , Fatores de Transcrição/genética , Raios Ultravioleta
17.
JCI Insight ; 4(24)2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31751318

RESUMO

As sufficient extracellular arginine is crucial for T cell function, depletion of extracellular arginine by elevated arginase 1 (Arg1) activity has emerged as a hallmark immunosuppressive mechanism. However, the potential cell-autonomous roles of arginases in T cells have remained unexplored. Here, we show that the arginase isoform expressed by T cells, the mitochondrial Arg2, is a cell-intrinsic regulator of CD8+ T cell activity. Both germline Arg2 deletion and adoptive transfer of Arg2-/- CD8+ T cells significantly reduced tumor growth in preclinical cancer models by enhancing CD8+ T cell activation, effector function, and persistence. Transcriptomic, proteomic, and high-dimensional flow cytometry characterization revealed a CD8+ T cell-intrinsic role of Arg2 in modulating T cell activation, antitumor cytoxicity, and memory formation, independently of extracellular arginine availability. Furthermore, specific deletion of Arg2 in CD8+ T cells strongly synergized with PD-1 blockade for the control of tumor growth and animal survival. These observations, coupled with the finding that pharmacologic arginase inhibition accelerates activation of ex vivo human T cells, unveil Arg2 as a potentially new therapeutic target for T cell-based cancer immunotherapies.


Assuntos
Arginase/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias Colorretais/imunologia , Melanoma Experimental/imunologia , Mitocôndrias/enzimologia , Animais , Arginase/genética , Arginina/metabolismo , Linfócitos T CD8-Positivos/enzimologia , Linhagem Celular Tumoral , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Citotoxicidade Imunológica , Feminino , Deleção de Genes , Humanos , Tolerância Imunológica/imunologia , Memória Imunológica/imunologia , Imunoterapia Adotiva/métodos , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Masculino , Melanoma Experimental/enzimologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/imunologia , Transplante de Neoplasias , Receptor de Morte Celular Programada 1/antagonistas & inibidores
18.
DNA Repair (Amst) ; 6(5): 602-14, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17208056

RESUMO

Mildly affected individuals from xeroderma pigmentosum complementation group G (XP-G) possess single amino acid substitutions in the XPG protein that adversely affects its 3' endonuclease function in nucleotide excision repair. More serious mutations in the XPG gene generate truncated or unstable XPG proteins and result in a particularly early and severe form of the combined XP/CS complex. Following UV irradiation, cells from such XP-G/CS patients enter apoptosis more readily than other DNA repair-deficient cells. Here, we explore the mechanisms by which UV triggers the apoptotic cell death program in XP-G and XP-G/CS primary fibroblasts. Activation of the CD95 signalling pathway occurs within minutes and it is the earliest detectable post-UV event in such cells. This is rapidly followed by activation of caspase-8 then caspase-3. Several hours later caspase-9 becomes activated and the mitochondrial membrane potential drops, but without any obvious prior release of cytochrome c. Although p53 accumulates in XPG-deficient cells after UV irradiation, use of RNA interference demonstrates that p53 is not required for their UV-induced apoptotic response. p53 ablation of wild-type fibroblasts reduces MDM2 mRNA levels, inhibits accumulation of the 90kDa/92kDa Mdm2 isoforms, and prevents the nuclear relocalisation of Mdm2 after UV treatment. The same post-UV effects occur in XPG-deficient cells that express normal p53 levels. These results emphasise the importance of the extrinsic apoptotic pathway and aberrant Mdm2 events for the severe UV-induced apoptosis of XPG-deficient primary fibroblasts. XP-G/CS cells constitutively overexpress the pro-apoptotic Bax protein and a long isoform of the E2F1 transcription factor that controls S phase entry, which may prime them to enter apoptosis very readily after UV irradiation.


Assuntos
Apoptose/efeitos da radiação , Caspase 3/metabolismo , Caspase 9/metabolismo , Proteínas de Ligação a DNA/fisiologia , Endonucleases/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta , Receptor fas/metabolismo , Células Cultivadas , Reparo do DNA , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Fibroblastos/fisiologia , Fibroblastos/efeitos da radiação , Humanos , Potencial da Membrana Mitocondrial , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/genética
19.
Mol Cell Biol ; 24(24): 10670-80, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15572672

RESUMO

XPG is the human endonuclease that cuts 3' to DNA lesions during nucleotide excision repair. Missense mutations in XPG can lead to xeroderma pigmentosum (XP), whereas truncated or unstable XPG proteins cause Cockayne syndrome (CS), normally yielding life spans of <7 years. One XP-G individual who had advanced XP/CS symptoms at 28 years has been identified. The genetic, biochemical, and cellular defects in this remarkable case provide insight into the onset of XP and CS, and they reveal a previously unrecognized property of XPG. Both of this individual's XPG alleles produce a severely truncated protein, but an infrequent alternative splice generates an XPG protein lacking seven internal amino acids, which can account for his very slight cellular UV resistance. Deletion of XPG amino acids 225 to 231 does not abolish structure-specific endonuclease activity. Instead, this region is essential for interaction with TFIIH and for the stable recruitment of XPG to sites of local UV damage after the prior recruitment of TFIIH. These results define a new functional domain of XPG, and they demonstrate that recruitment of DNA repair proteins to sites of damage does not necessarily lead to productive repair reactions. This observation has potential implications that extend beyond nucleotide excision repair.


Assuntos
Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição TFII/metabolismo , Raios Ultravioleta , Processamento Alternativo , Sequência de Aminoácidos , Linhagem Celular , Linhagem Celular Transformada , Transformação Celular Viral , Análise Mutacional de DNA , Reparo do DNA , Proteínas de Ligação a DNA/genética , Endonucleases/análise , Endonucleases/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Técnica Indireta de Fluorescência para Anticorpo , Mutação da Fase de Leitura , Humanos , Immunoblotting , Lentivirus/genética , Longevidade , Masculino , Microscopia de Fluorescência , Proteínas Nucleares , Testes de Precipitina , Estrutura Terciária de Proteína , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Fator de Transcrição TFIIH , Fatores de Transcrição , Xeroderma Pigmentoso/diagnóstico , Xeroderma Pigmentoso/genética , Xeroderma Pigmentoso/metabolismo , Xeroderma Pigmentoso/patologia
20.
Mol Biol Cell ; 13(5): 1626-40, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12006658

RESUMO

Eukaryotic translation initiation factor 3 (eIF3) is a multisubunit complex that plays a central role in translation initiation. We show that fission yeast Sum1, which is structurally related to known eIF3 subunits in other species, is essential for translation initiation, whereas its overexpression results in reduced global translation. Sum1 is associated with the 40S ribosome and interacts stably with Int6, an eIF3 component, in vivo, suggesting that Sum1 is a component of the eIF3 complex. Sum1 is cytoplasmic under normal growth conditions. Surprisingly, Sum1 is rapidly relocalized to cytoplasmic foci after osmotic and thermal stress. Int6 and p116, another putative eIF3 subunit, behave similarly, suggesting that eIF3 is a dynamic complex. These cytoplasmic foci, which additionally comprise eIF4E and RNA components, may function as translation centers during environmental stress. After heat shock, Sum1 additionally colocalizes stably with the 26S proteasome at the nuclear periphery. The relationship between Sum1 and the 26S proteasome was further investigated, and we find cytoplasmic Sum1 localization to be dependent on the 26S proteasome. Furthermore, Sum1 interacts with the Mts2 and Mts4 components of the 26S proteasome. These data indicate a functional link between components of the structurally related eIF3 translation initiation and 26S proteasome complexes.


Assuntos
Fator de Iniciação 3 em Eucariotos/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Nucleares/metabolismo , Peptídeo Hidrolases/metabolismo , Complexo de Endopeptidases do Proteassoma , Proteínas de Saccharomyces cerevisiae , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Transporte/metabolismo , Endopeptidases/metabolismo , Proteínas Fúngicas/genética , Proteínas de Fluorescência Verde , Temperatura Alta , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Substâncias Macromoleculares , Proteínas Nucleares/genética , Pressão Osmótica , Biossíntese de Proteínas , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras , Schizosaccharomyces/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA