Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 38(1): 66-78, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23177319

RESUMO

Suppressors of cytokine signaling (SOCS) are important regulators of lipopolysaccharide (LPS) and cytokine responses but their role in macrophage polarization is unknown. We have shown here that myeloid-restricted Socs3 deletion (Socs3(Lyz2cre)) resulted in resistance to LPS-induced endotoxic shock, whereas Socs2(-/-) mice were highly susceptible. We observed striking bias toward M2-like macrophages in Socs3(Lyz2cre) mice, whereas the M1-like population was enriched in Socs2(-/-) mice. Adoptive transfer experiments showed that responses to endotoxic shock and polymicrobial sepsis were transferable and macrophage dependent. Critically, this dichotomous response was associated with enhanced regulatory T (Treg) cell recruitment by Socs3(Lyz2cre) cells, whereas Treg cell recruitment was absent in the presence of Socs2(-/-) macrophages. In addition, altered polarization coincided with enhanced interferon-gamma (IFN-γ)-induced signal transducer and activator of transcription-1 (STAT1) activation in Socs2(-/-) macrophages and enhanced interleukin-4 (IL-4) plus IL-13-induced STAT6 phosphorylation in Socs3(Lyz2cre) macrophages. SOCS, therefore, are essential controllers of macrophage polarization, regulating inflammatory responses.


Assuntos
Polaridade Celular/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Transferência Adotiva , Animais , Regulação da Expressão Gênica , Interleucina-10/imunologia , Interleucina-10/metabolismo , Macrófagos/transplante , Camundongos , Fatores de Transcrição STAT/metabolismo , Sepse/genética , Sepse/imunologia , Sepse/prevenção & controle , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transplante Isogênico
2.
Cytotherapy ; 23(2): 131-136, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33309258

RESUMO

BACKGROUND AIMS: To investigate the feasibility of using CD4 + T cells genetically modified to express an allo-HLA directed CAR and FOXP3 to suppress T cell proliferation and cytokine secretion in GvHD. METHODS: Human CD4+ T cells from A*02:01 negative donors were transduced to express A*02 CAR and FOXP3 and co-cultured in mixed lymphocyte reaction assays to demonstrate T cell suppression. A*02- CAR/FOXP CD4+ T cells were then injected into mice engrafted with allogeneic T cells in a GvHD mouse model. RESULTS: CD4+ T cells genetically modified to express allo-HLA-directed CAR and FOXP3 proliferate rapidly, downregulate CD127 and interferon-γ, express high CD25 and Helios and convert to a stable antigen-dependent suppressive phenotype. In mixed lymphocyte reaction assays, these cells potently suppressed T-cell proliferation and secreted IL-10. In a graft-versus-host disease model, A*02-CAR/FOXP3 CD4+ T cells outperformed polyclonal Tregs by reducing liver and lung inflammation, inhibiting pro-inflammatory cytokine production and limiting grafted CD3+ T-cell expansion. CONCLUSIONS: CD4 + T cells expressing allo-antigen directed HLA-specific CAR and FOXP3 act as potent, specific and stable suppressors of inflammation that out-perform their Treg counterparts both in vitro and in vivo.


Assuntos
Linfócitos T CD4-Positivos , Doença Enxerto-Hospedeiro , Receptores de Antígenos Quiméricos , Linfócitos T Reguladores , Animais , Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/genética , Antígenos HLA-A , Humanos , Camundongos , Fenótipo , Linfócitos T Reguladores/imunologia
3.
PLoS Genet ; 13(3): e1006659, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28273074

RESUMO

IL-33 is a tissue-derived cytokine that induces and amplifies eosinophilic inflammation and has emerged as a promising new drug target for asthma and allergic disease. Common variants at IL33 and IL1RL1, encoding the IL-33 receptor ST2, associate with eosinophil counts and asthma. Through whole-genome sequencing and imputation into the Icelandic population, we found a rare variant in IL33 (NM_001199640:exon7:c.487-1G>C (rs146597587-C), allele frequency = 0.65%) that disrupts a canonical splice acceptor site before the last coding exon. It is also found at low frequency in European populations. rs146597587-C associates with lower eosinophil counts (ß = -0.21 SD, P = 2.5×10-16, N = 103,104), and reduced risk of asthma in Europeans (OR = 0.47; 95%CI: 0.32, 0.70, P = 1.8×10-4, N cases = 6,465, N controls = 302,977). Heterozygotes have about 40% lower total IL33 mRNA expression than non-carriers and allele-specific analysis based on RNA sequencing and phased genotypes shows that only 20% of the total expression is from the mutated chromosome. In half of those transcripts the mutation causes retention of the last intron, predicted to result in a premature stop codon that leads to truncation of 66 amino acids. The truncated IL-33 has normal intracellular localization but neither binds IL-33R/ST2 nor activates ST2-expressing cells. Together these data demonstrate that rs146597587-C is a loss of function mutation and support the hypothesis that IL-33 haploinsufficiency protects against asthma.


Assuntos
Asma/genética , Eosinófilos/metabolismo , Interleucina-33/genética , Mutação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Processamento Alternativo , Animais , Sítios de Ligação , Bioensaio , Criança , Pré-Escolar , Dinamarca , Feminino , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Heterozigoto , Humanos , Islândia , Lactente , Recém-Nascido , Íntrons , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Países Baixos , Adulto Jovem
4.
J Immunol ; 199(2): 707-717, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28615416

RESUMO

Atopic dermatitis (AD) is a common inflammatory skin disease affecting up to 20% of children and 3% of adults worldwide and is associated with dysregulation of the skin barrier. Although type 2 responses are implicated in AD, emerging evidence indicates a potential role for the IL-17A signaling axis in AD pathogenesis. In this study we show that in the filaggrin mutant mouse model of spontaneous AD, IL-17RA deficiency (Il17ra-/- ) resulted in severe exacerbation of skin inflammation. Interestingly, Il17ra-/- mice without the filaggrin mutation also developed spontaneous progressive skin inflammation with eosinophilia, as well as increased levels of thymic stromal lymphopoietin (TSLP) and IL-5 in the skin. Il17ra-/- mice have a defective skin barrier with altered filaggrin expression. The barrier dysregulation and spontaneous skin inflammation in Il17ra-/- mice was dependent on TSLP, but not the other alarmins IL-25 and IL-33. The associated skin inflammation was mediated by IL-5-expressing pathogenic effector Th2 cells and was independent of TCRγδ T cells and IL-22. An absence of IL-17RA in nonhematopoietic cells, but not in the hematopoietic cells, was required for the development of spontaneous skin inflammation. Skin microbiome dysbiosis developed in the absence of IL-17RA, with antibiotic intervention resulting in significant amelioration of skin inflammation and reductions in skin-infiltrating pathogenic effector Th2 cells and TSLP. This study describes a previously unappreciated protective role for IL-17RA signaling in regulation of the skin barrier and maintenance of skin immune homeostasis.


Assuntos
Dermatite Atópica/imunologia , Receptores de Interleucina-17/imunologia , Receptores de Interleucina-17/metabolismo , Pele/crescimento & desenvolvimento , Pele/patologia , Animais , Citocinas/imunologia , Dermatite Atópica/patologia , Modelos Animais de Doenças , Disbiose , Eosinofilia/imunologia , Proteínas Filagrinas , Regulação da Expressão Gênica , Homeostase , Interleucina-33/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Interleucinas/genética , Interleucinas/imunologia , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/genética , Camundongos , Microbiota , Mutação , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Interleucina-17/deficiência , Receptores de Interleucina-17/genética , Transdução de Sinais , Pele/imunologia , Pele/microbiologia , Células Th2/imunologia , Linfopoietina do Estroma do Timo , Interleucina 22
5.
Eur Respir J ; 48(3): 715-25, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27338192

RESUMO

Severe asthma represents a major unmet clinical need. Eosinophilic inflammation persists in the airways of many patients with uncontrolled asthma, despite high-dose inhaled corticosteroid therapy. Suppressors of cytokine signalling (SOCS) are a family of molecules involved in the regulation of cytokine signalling via inhibition of the Janus kinase-signal transducers and activators of transcription pathway. We examined SOCS expression in the airways of asthma patients and investigated whether this is associated with persistent eosinophilia.Healthy controls, mild/moderate asthmatics and severe asthmatics were studied. Whole genome expression profiling, quantitative PCR and immunohistochemical analysis were used to examine expression of SOCS1, SOCS2 and SOCS3 in bronchial biopsies. Bronchial epithelial cells were utilised to examine the role of SOCS1 in regulating interleukin (IL)-13 signalling in vitroSOCS1 gene expression was significantly lower in the airways of severe asthmatics compared with mild/moderate asthmatics, and was inversely associated with airway eosinophilia and other measures of T-helper type 2 (Th2) inflammation. Immunohistochemistry demonstrated SOCS1 was predominantly localised to the bronchial epithelium. SOCS1 overexpression inhibited IL-13-mediated chemokine ligand (CCL) 26 (eotaxin-3) mRNA expression in bronchial epithelial cells.Severe asthma patients with persistent airway eosinophilia and Th2 inflammation have reduced airway epithelial SOCS1 expression. SOCS1 inhibits epithelial IL-13 signalling, supporting its key role in regulating Th2-driven eosinophilia in severe asthma.


Assuntos
Asma/metabolismo , Células Epiteliais/metabolismo , Interleucina-13/metabolismo , Eosinofilia Pulmonar/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Adulto , Asma/tratamento farmacológico , Biópsia , Brônquios/metabolismo , Broncoscopia , Estudos de Casos e Controles , Linhagem Celular , Quimiocina CCL26/metabolismo , Estudos de Coortes , Feminino , Perfilação da Expressão Gênica , Humanos , Inflamação , Masculino , Pessoa de Meia-Idade , Eosinofilia Pulmonar/tratamento farmacológico , Mucosa Respiratória/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Células Th2/citologia , Adulto Jovem
6.
J Immunol ; 190(7): 3235-45, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23455506

RESUMO

Suppressor of cytokine signaling (SOCS) proteins are key regulators of CD4(+) T cell differentiation, and in particular, we have recently shown that SOCS2 inhibits the development of Th2 cells and allergic immune responses. Interestingly, transcriptome analyses have identified SOCS2 as being preferentially expressed in both natural regulatory T cells (Tregs) and inducible Tregs (iTregs); however, the role of SOCS2 in Foxp3(+) Treg function or development has not been fully elucidated. In this study, we show that despite having no effect on natural Treg development or function, SOCS2 is highly expressed in iTregs and required for the stable expression of Foxp3 in iTregs in vitro and in vivo. Indeed, SOCS2-deficient CD4(+) T cells upregulated Foxp3 following in vitro TGF-ß stimulation, but failed to maintain stable expression of Foxp3. Moreover, in vivo generation of iTregs following OVA feeding was impaired in the absence of SOCS2 and could be rescued in the presence of IL-4 neutralizing Ab. Following IL-4 stimulation, SOCS2-deficient Foxp3(+) iTregs secreted elevated IFN-γ and IL-13 levels and displayed enhanced STAT6 phosphorylation. Therefore, we propose that SOCS2 regulates iTreg stability by downregulating IL-4 signaling. Moreover, SOCS2 is essential to maintain the anti-inflammatory phenotype of iTregs by preventing the secretion of proinflammatory cytokines. Collectively, these results suggest that SOCS2 may prevent IL-4-induced Foxp3(+) iTreg instability. Foxp3(+) iTregs are key regulators of immune responses at mucosal surfaces; therefore, this dual role of SOCS2 in both Th2 and Foxp3(+) iTregs reinforces SOCS2 as a potential therapeutic target for Th2-biased diseases.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Interleucina-4/farmacologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Fator de Transcrição STAT6/metabolismo , Proteínas Supressoras da Sinalização de Citocina/deficiência , Proteínas Supressoras da Sinalização de Citocina/genética , Linfócitos T Reguladores/efeitos dos fármacos
7.
Biochem J ; 457(2): 289-300, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24134311

RESUMO

Processing of the 'CaaX' motif found on the C-termini of many proteins, including the proto-oncogene Ras, requires the ER (endoplasmic reticulum)-resident protease RCE1 (Ras-converting enzyme 1) and is necessary for the proper localization and function of many of these 'CaaX' proteins. In the present paper, we report that several mammalian species have a novel isoform (isoform 2) of RCE1 resulting from an alternate splice site and producing an N-terminally truncated protein. We demonstrate that both RCE1 isoform 1 and the newly identified isoform 2 are required to reinstate proper H-Ras processing and thus plasma membrane localization in RCE1-null cells. In addition, we show that the deubiquitinating enzyme USP17 (ubiquitin-specific protease 17), previously shown to modulate RCE1 activity, can regulate the abundance and localization of isoform 2. Furthermore, we show that isoform 2 is ubiquitinated on Lys43 and deubiquitinated by USP17. Collectively, the findings of the present study indicate that RCE1 isoform 2 is required for proper 'CaaX' processing and that USP17 can regulate this via its modulation of RCE1 isoform 2 ubiquitination.


Assuntos
Membrana Celular/metabolismo , Endopeptidases/metabolismo , Endopeptidases/fisiologia , Genes ras/fisiologia , Membrana Celular/química , Células HEK293 , Células HeLa , Humanos , Isoformas de Proteínas/metabolismo , Proto-Oncogene Mas
8.
J Immunol ; 189(5): 2079-83, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22837488

RESUMO

GM-CSF is a potent proinflammatory cytokine that plays a pathogenic role in the CNS inflammatory disease experimental autoimmune encephalomyelitis. As IL-27 alleviates experimental autoimmune encephalomyelitis, we hypothesized that IL-27 suppresses GM-CSF expression by T cells. We found that IL-27 suppressed GM-CSF expression in CD4+ and CD8+ T cells in splenocyte and purified T cell cultures. IL-27 suppressed GM-CSF in Th1, but not Th17, cells. IL-27 also suppressed GM-CSF expression by human T cells in nonpolarized and Th1- but not Th17-polarized PBMC cultures. In vivo, IL-27p28 deficiency resulted in increased GM-CSF expression by CNS-infiltrating T cells during Toxoplasma gondii infection. Although in vitro suppression of GM-CSF by IL-27 was independent of IL-2 suppression, IL-10 upregulation, or SOCS3 signaling, we observed that IL-27-driven suppression of GM-CSF was STAT1 dependent. Our findings demonstrate that IL-27 is a robust negative regulator of GM-CSF expression in T cells, which likely inhibits T cell pathogenicity in CNS inflammation.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Tolerância Imunológica , Interleucina-17/fisiologia , Subpopulações de Linfócitos T/imunologia , Animais , Polaridade Celular/genética , Polaridade Celular/imunologia , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Tolerância Imunológica/genética , Mediadores da Inflamação/farmacologia , Mediadores da Inflamação/fisiologia , Interleucina-17/farmacologia , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Toxoplasmose/imunologia , Toxoplasmose/patologia
9.
Int J Cancer ; 133(9): 2102-12, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23629809

RESUMO

Recent murine studies have demonstrated that tumor-associated macrophages in the tumor microenvironment are a key source of the pro-tumorigenic cysteine protease, cathepsin S. We now show in a syngeneic colorectal carcinoma murine model that both tumor and tumor-associated cells contribute cathepsin S to promote neovascularization and tumor growth. Cathepsin S depleted and control colorectal MC38 tumor cell lines were propagated in both wild type C57Bl/6 and cathepsin S null mice to provide stratified depletion of the protease from either the tumor, tumor-associated host cells, or both. Parallel analysis of these conditions showed that deletion of cathepsin S inhibited tumor growth and development, and revealed a clear contribution of both tumor and tumor-associated cell derived cathepsin S. The most significant impact on tumor development was obtained when the protease was depleted from both sources. Further characterization revealed that the loss of cathepsin S led to impaired tumor vascularization, which was complemented by a reduction in proliferation and increased apoptosis, consistent with reduced tumor growth. Analysis of cell types showed that in addition to the tumor cells, tumor-associated macrophages and endothelial cells can produce cathepsin S within the microenvironment. Taken together, these findings clearly highlight a manner by which tumor-associated cells can positively contribute to developing tumors and highlight cathepsin S as a therapeutic target in cancer.


Assuntos
Carcinoma Pulmonar de Lewis/patologia , Catepsinas/fisiologia , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/patologia , Melanoma Experimental/patologia , Neovascularização Patológica , Animais , Apoptose , Western Blotting , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/genética , Adesão Celular , Ciclo Celular , Células Cultivadas , Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/genética , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Macrófagos/citologia , Macrófagos/metabolismo , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Microambiente Tumoral
10.
Sci Adv ; 9(25): eade7890, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37352349

RESUMO

Peptides from degradation of intracellular proteins are continuously displayed by major histocompatibility complex (MHC) class I. To better understand origins of these peptides, we performed a comprehensive census of the class I peptide repertoire in the presence and absence of ubiquitin-proteasome system (UPS) activity upon developing optimized methodology to enrich for and quantify these peptides. Whereas most class I peptides are dependent on the UPS for their generation, a surprising 30%, enriched in peptides of mitochondrial origin, appears independent of the UPS. A further ~10% of peptides were found to be dependent on the proteasome but independent of ubiquitination for their generation. Notably, clinically achievable partial inhibition of the proteasome resulted in display of atypical peptides. Our results suggest that generation of MHC class I•peptide complexes is more complex than previously recognized, with UPS-dependent and UPS-independent components; paradoxically, alternative protein degradation pathways also generate class I peptides when canonical pathways are impaired.


Assuntos
Apresentação de Antígeno , Complexo de Endopeptidases do Proteassoma , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Antígenos de Histocompatibilidade Classe I/metabolismo , Peptídeos/metabolismo , Ubiquitina/metabolismo
11.
Immunology ; 135(2): 101-11, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22044277

RESUMO

Suppressors of cytokine signalling (SOCS) proteins are induced in responses to many stimuli and by binding to cytokine receptors and associated janus kinase (JAK) proteins, directly regulate the activation of the signal transducers and activators of transcription (STATs). STAT proteins regulate the expression of many genes required for the differentiation of various CD4(+) T helper cell lineages, and there is now accumulating evidence that SOCS also play essential roles in the regulation and maintenance of CD4(+) T-cell polarization. As it is now clear that CD4(+) T cells are more plastic than initially thought, it is of particular importance to understand the molecular mechanisms regulating CD4(+) T-cell differentiation. Here we review the current understanding of how STATs and SOCS act in concert to influence the polarization of CD4(+) T cells and highlight the relevance of this in disease.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Linhagem da Célula , Humanos , Fatores de Transcrição STAT/imunologia , Fatores de Transcrição STAT/metabolismo , Proteínas Supressoras da Sinalização de Citocina/imunologia
12.
J Immunol ; 184(12): 6575-84, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20483741

RESUMO

The monomeric GTPase Rap1 controls functional activation of beta2 integrins in leukocytes. In this article, we describe a novel mechanism by which the chemoattractant fMLP activates Rap1 and inside-out signaling of beta2 integrins. We found that fMLP-induced activation of Rap1 in human polymorphonuclear leukocytes or neutrophils and differentiated PLB-985 cells was blocked by inhibitors of the NO/guanosine-3',5'-cyclic monophosphate-dependent protein kinase (cGKI) pathway [N-(3-(aminomethyl)benzyl)acetamidine, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, DT-3 peptide, 8-(4-chlorophenylthio)guanosine 3',5'-cyclic monophosphothioate, Rp-isomer triethylammonium salt-guanosine-3',5'-cyclic monophosphate], indicating that the downstream signaling events in Rap1 activation involve the production of NO and guanosine-3',5'-cyclic monophosphate, as well as the activation of cGKI. Silencing the expression of vasodilator-stimulated phosphoprotein (VASP), a substrate of cGKI, in resting PLB-985 cells or mice neutrophils led to constitutive activation of Rap1. In parallel, silencing VASP in differentiated PLB-985 cells led to recruitment of C3G, a guanine nucleotide exchange factor for Rap1, to the plasma membrane. Expression of murine GFP-tagged phosphodeficient VASP Ser235Ala mutant (murine serine 235 of VASP corresponds to human serine 239) in PLB-985 cells blunted fMLP-induced translocation of C3G to the membrane and activation of Rap1. Thus, bacterial fMLP triggers cGKI-dependent phosphorylation of human VASP on serine 239 and, thereby, controls membrane recruitment of C3G, which is required for activation of Rap1 and beta2 integrin-dependent antibacterial functions of neutrophils.


Assuntos
Antígenos CD18/imunologia , Moléculas de Adesão Celular/imunologia , Proteínas dos Microfilamentos/imunologia , Neutrófilos/imunologia , Fosfoproteínas/imunologia , Transdução de Sinais/imunologia , Animais , Proteínas de Bactérias/imunologia , Western Blotting , Antígenos CD18/metabolismo , Moléculas de Adesão Celular/metabolismo , Fator 2 de Liberação do Nucleotídeo Guanina/imunologia , Fator 2 de Liberação do Nucleotídeo Guanina/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Neutrófilos/metabolismo , Fosfoproteínas/metabolismo , Transporte Proteico/fisiologia , Proteínas rap1 de Ligação ao GTP/imunologia , Proteínas rap1 de Ligação ao GTP/metabolismo
13.
J Biol Chem ; 285(16): 12028-36, 2010 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-20147298

RESUMO

The proto-oncogenic Ras isoforms (H, N, and K) have a C-terminal CAAX motif and undergo the same post-translational processing steps, although they traffic to the plasma membrane through different routes. Previously, we have shown that overexpression of the deubiquitinating enzyme USP17 inhibits H-Ras localization to the plasma membrane. Now we report that whereas H-Ras and N-Ras were unable to localize to the plasma membrane in the presence of USP17, K-Ras4b localization was unaffected. EGF stimulation was unable to induce N-Ras membrane localization in USP17-expressing cells. In addition, N-Ras activity and downstream signaling through the MAPK MEK/ERK and PI3K/JNK pathways were blunted. However, we still detected abundant N-Ras localization at the ER and Golgi in USP17-expressing cells. Collectively, our data showed that the deubiquitinating enzyme USP17 blocks EGF-induced N-Ras membrane trafficking and activation, but left K-Ras unaffected.


Assuntos
Endopeptidases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transporte Biológico Ativo/efeitos dos fármacos , Brefeldina A/farmacologia , Membrana Celular/metabolismo , Endopeptidases/genética , Retículo Endoplasmático/metabolismo , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Transfecção , Proteases Específicas de Ubiquitina
14.
Mol Cancer ; 10: 147, 2011 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-22168338

RESUMO

BACKGROUND: Proteolytic enzymes have been implicated in driving tumor progression by means of their cancer cell microenvironment activity where they promote proliferation, differentiation, apoptosis, migration, and invasion. Therapeutic strategies have focused on attenuating their activity using small molecule inhibitors, but the association of proteases with the cell surface during cancer progression opens up the possibility of targeting these using antibody dependent cellular cytotoxicity (ADCC). Cathepsin S is a lysosomal cysteine protease that promotes the growth and invasion of tumour and endothelial cells during cancer progression. Our analysis of colorectal cancer patient biopsies shows that cathepsin S associates with the cell membrane indicating a potential for ADCC targeting. RESULTS: Here we report the cell surface characterization of cathepsin S and the development of a humanized antibody (Fsn0503h) with immune effector function and a stable in vivo half-life of 274 hours. Cathepsin S is expressed on the surface of tumor cells representative of colorectal and pancreatic cancer (23%-79% positive expression). Furthermore the binding of Fsn0503h to surface associated cathepsin S results in natural killer (NK) cell targeted tumor killing. In a colorectal cancer model Fsn0503h elicits a 22% cytotoxic effect. CONCLUSIONS: This data highlights the potential to target cell surface associated enzymes, such as cathepsin S, as therapeutic targets using antibodies capable of elicitingADCC in tumor cells.


Assuntos
Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Catepsinas/imunologia , Citotoxicidade Imunológica , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacocinética , Catepsinas/química , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Feminino , Citometria de Fluxo , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Masculino , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Ratos , Ratos Sprague-Dawley
15.
J Immunol ; 183(12): 7703-9, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19933851

RESUMO

Although production of cytokines by TLR is essential for viral and bacterial clearance, overproduction can be detrimental, thus controlling these responses is essential. CD33-related sialic acid binding Ig-like lectin receptors (Siglecs) have been implicated in the control of leukocyte responses. In this study, we report that murine Siglec-E is induced by TLRs in a MyD88-specific manner, is tyrosine phosphorylated following LPS stimulation, and negatively regulates TLR responses. Specifically, we demonstrate the Siglec-E expression inhibits TLR-induced NF-kappaB and more importantly, the induction of the antiviral cytokines IFN-beta and RANTES. Siglec-E mediates its inhibitory effects on TIR domain containing adaptor inducing IFN-beta (TRIF)-dependent cytokine production via recruitment of the tyrosine [corrected] phosphatase SHP2 and subsequent inhibition of TBK1 activity as evidenced by enhanced TBK1 phosphorylation in cells following knockdown of Siglec-E expression. Taken together, our results demonstrate a novel role for Siglec-E in controlling the antiviral response to TLRs and thus helping to maintain a healthy cytokine balance following infection.


Assuntos
Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos B/biossíntese , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Regulação para Baixo/imunologia , Lipopolissacarídeos/farmacologia , Receptores Toll-Like/antagonistas & inibidores , Receptores Toll-Like/fisiologia , Regulação para Cima/imunologia , Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Antígenos CD/metabolismo , Antígenos CD/fisiologia , Antígenos de Diferenciação de Linfócitos B/metabolismo , Antígenos de Diferenciação de Linfócitos B/fisiologia , Linhagem Celular , Linhagem Celular Transformada , Citocinas/genética , Regulação para Baixo/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fosforilação/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
16.
Nat Med ; 9(8): 1047-54, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12847520

RESUMO

Members of the suppressor of cytokine signaling (SOCS) family are involved in the pathogenesis of many inflammatory diseases. SOCS-3 is predominantly expressed in T-helper type 2 (T(H)2) cells, but its role in T(H)2-related allergic diseases remains to be investigated. In this study we provide a strong correlation between SOCS-3 expression and the pathology of asthma and atopic dermatitis, as well as serum IgE levels in allergic human patients. SOCS-3 transgenic mice showed increased T(H)2 responses and multiple pathological features characteristic of asthma in an airway hypersensitivity model system. In contrast, dominant-negative mutant SOCS-3 transgenic mice, as well as mice with a heterozygous deletion of Socs3, had decreased T(H)2 development. These data indicate that SOCS-3 has an important role in regulating the onset and maintenance of T(H)2-mediated allergic immune disease, and suggest that SOCS-3 may be a new therapeutic target for the development of antiallergic drugs.


Assuntos
Asma/imunologia , Dermatite Atópica/imunologia , Hipersensibilidade/imunologia , Proteínas/metabolismo , Proteínas Repressoras , Células Th2/imunologia , Fatores de Transcrição , Animais , Hiper-Reatividade Brônquica/imunologia , Testes de Provocação Brônquica , Proteínas de Ligação a DNA/metabolismo , Humanos , Hipersensibilidade/metabolismo , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas/genética , Fator de Transcrição STAT4 , Transdução de Sinais/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Células Th2/fisiologia , Transativadores/metabolismo
17.
BMC Genomics ; 11: 250, 2010 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-20403174

RESUMO

BACKGROUND: The DUB/USP17 subfamily of deubiquitinating enzymes were originally identified as immediate early genes induced in response to cytokine stimulation in mice (DUB-1, DUB-1A, DUB-2, DUB-2A). Subsequently we have identified a number of human family members and shown that one of these (DUB-3) is also cytokine inducible. We originally showed that constitutive expression of DUB-3 can block cell proliferation and more recently we have demonstrated that this is due to its regulation of the ubiquitination and activity of the 'CAAX' box protease RCE1. RESULTS: Here we demonstrate that the human DUB/USP17 family members are found on both chromosome 4p16.1, within a block of tandem repeats, and on chromosome 8p23.1, embedded within the copy number variable beta-defensin cluster. In addition, we show that the multiple genes observed in humans and other distantly related mammals have arisen due to the independent expansion of an ancestral sequence within each species. However, it is also apparent when sequences from humans and the more closely related chimpanzee are compared, that duplication events have taken place prior to these species separating. CONCLUSIONS: The observation that the DUB/USP17 genes, which can influence cell growth and survival, have evolved from an unstable ancestral sequence which has undergone multiple and varied duplications in the species examined marks this as a unique family. In addition, their presence within the beta-defensin repeat raises the question whether they may contribute to the influence of this repeat on immune related conditions.


Assuntos
Variações do Número de Cópias de DNA , Endopeptidases/genética , beta-Defensinas/genética , Animais , Cromossomos Humanos Par 4 , Cromossomos Humanos Par 8 , Humanos , Camundongos , Família Multigênica , Filogenia , Ratos , Sequências de Repetição em Tandem , Proteases Específicas de Ubiquitina
18.
J Exp Med ; 197(4): 425-36, 2003 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-12591901

RESUMO

Suppressor of cytokine signaling (SOCS)3 has been characterized as a negative feedback regulator in cytokine-mediated Janus kinase signal transducer and activator of transcription signaling. However, this study shows that T cells from transgenic mice expressing SOCS3 exhibit a significant reduction in interleukin (IL)-2 production induced by T cell receptor cross-linking when T cells are costimulated with CD28. Decreased protein expression in SOCS3(+/-) mice enhanced CD28-mediated IL-2 production, clearly indicating the correlation between expression level of SOCS3 and IL-2 production ability. The SOCS3 protein interacted with phosphorylated CD28 through its SH2 domain but not the kinase inhibitory region. In addition, a point mutation in the SOCS3 SH2 domain attenuated the inhibition of CD28 function in IL-2 promoter activation. Committed T helper (Th)2 cells exclusively expressed SOCS3 and production of Th2 cytokines, such as IL-4 and IL-5, was much less dependent on CD28 costimulation compared with interferon gamma and IL-2 production in Th1 cells. Consistent with this notion, the expression level of SOCS3 in early T cell activation influenced the ability of IL-2 production induced by CD28 costimulation. Therefore, the SOCS3 may play an alternative role in prohibiting excessive progression of CD28-mediated IL-2 production.


Assuntos
Antígenos CD28/fisiologia , Interleucina-2/biossíntese , Proteínas/fisiologia , Proteínas Repressoras , Fatores de Transcrição , Animais , Humanos , Interleucina-2/genética , Interleucina-4/farmacologia , Células Jurkat , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Regiões Promotoras Genéticas , Receptores de Antígenos de Linfócitos T/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Linfócitos T/imunologia , Tirosina/metabolismo , Domínios de Homologia de src
19.
J Leukoc Biol ; 85(2): 289-97, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18971287

RESUMO

The chemokine eotaxin/CCL11 is an important mediator of leukocyte migration, but its effect on inflammatory cytokine signaling has not been explored. In this study, we find that CCL11 induces suppressor of cytokine signaling (SOCS)1 and SOCS3 expression in murine macrophages, human monocytes, and dendritic cells (DCs). We also discover that CCL11 inhibits GM-CSF-mediated STAT5 activation and IL-4-induced STAT6 activation in a range of hematopoietic cells. This blockade of cytokine signaling by CCL11 results in reduced differentiation and endocytic ability of DCs, implicating CCL11-induced SOCS as mediators of chemotactic inflammatory control. These findings demonstrate cross-talk between chemokine and cytokine responses, suggesting that myeloid cells tracking to the inflammatory site do not differentiate in the presence of this chemokine, revealing another role for SOCS in inflammatory regulation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Quimiocina CCL11/farmacologia , Células Dendríticas/citologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Sistema Hematopoético/citologia , Interleucina-4/farmacologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Endocitose/efeitos dos fármacos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Sistema Hematopoético/efeitos dos fármacos , Sistema Hematopoético/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética
20.
NPJ Vaccines ; 5(1): 106, 2020 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-33298945

RESUMO

Cancer vaccines using synthetic long peptides (SLP) targeting tumor antigens have been tested in the clinic but the outcomes have been unimpressive, perhaps because these peptides elicit predominantly CD4+ T cell responses. We hypothesized that enhanced delivery of peptide antigens to, and uptake in, secondary lymphoid tissues should elicit more robust CD8+ and CD4+ T cell responses and improved anti-tumor responses. Here, we have designed SLP-containing cationic lipoplexes (SLP-Lpx) that improve delivery of peptides to myeloid cells in the spleen and lymphatics. Using the G12D KRAS mutations as neoantigens, we found that vaccination of mice with naked synthetic peptides harboring the G12D mutation with CpG adjuvant stimulated mainly CD4+ T cell responses with limited tumor growth inhibition. On the other hand, immunization with SLP-Lpx stimulated both CD4+ and CD8+ T cells and suppressed tumor growth in a CD8+ T cell-dependent manner. Combination of the SLP-Lpx vaccines with a checkpoint inhibitor led to profound growth suppression of established tumors. These studies suggest that preferential targeting of peptides derived from neoantigens to the spleen via lipoplexes elicits potent CD4+ and CD8+ T cell responses that inhibit tumor growth.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA