Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Immunity ; 57(5): 1019-1036.e9, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38677292

RESUMO

Group 3 innate lymphoid cells (ILC3) are the major subset of gut-resident ILC with essential roles in infections and tissue repair, but how they adapt to the gut environment to maintain tissue residency is unclear. We report that Tox2 is critical for gut ILC3 maintenance and function. Gut ILC3 highly expressed Tox2, and depletion of Tox2 markedly decreased ILC3 in gut but not at central sites, resulting in defective control of Citrobacter rodentium infection. Single-cell transcriptional profiling revealed decreased expression of Hexokinase-2 in Tox2-deficient gut ILC3. Consistent with the requirement for hexokinases in glycolysis, Tox2-/- ILC3 displayed decreased ability to utilize glycolysis for protein translation. Ectopic expression of Hexokinase-2 rescued Tox2-/- gut ILC3 defects. Hypoxia and interleukin (IL)-17A each induced Tox2 expression in ILC3, suggesting a mechanism by which ILC3 adjusts to fluctuating environments by programming glycolytic metabolism. Our results reveal the requirement for Tox2 to support the metabolic adaptation of ILC3 within the gastrointestinal tract.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae , Glicólise , Imunidade Inata , Linfócitos , Camundongos Knockout , Animais , Camundongos , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Transativadores/metabolismo , Transativadores/genética , Hexoquinase/metabolismo , Hexoquinase/genética , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Interleucina-17/metabolismo , Adaptação Fisiológica/imunologia
2.
Nature ; 626(8001): 1102-1107, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38355795

RESUMO

Plasma cells produce large quantities of antibodies and so play essential roles in immune protection1. Plasma cells, including a long-lived subset, reside in the bone marrow where they depend on poorly defined microenvironment-linked survival signals1. We show that bone marrow plasma cells use the ligand-gated purinergic ion channel P2RX4 to sense extracellular ATP released by bone marrow osteoblasts through the gap-junction protein pannexin 3 (PANX3). Mutation of Panx3 or P2rx4 each caused decreased serum antibodies and selective loss of bone marrow plasma cells. Compared to their wild-type counterparts, PANX3-null osteoblasts secreted less extracellular ATP and failed to support plasma cells in vitro. The P2RX4-specific inhibitor 5-BDBD abrogated the impact of extracellular ATP on bone marrow plasma cells in vitro, depleted bone marrow plasma cells in vivo and reduced pre-induced antigen-specific serum antibody titre with little posttreatment rebound. P2RX4 blockade also reduced autoantibody titre and kidney disease in two mouse models of humoral autoimmunity. P2RX4 promotes plasma cell survival by regulating endoplasmic reticulum homeostasis, as short-term P2RX4 blockade caused accumulation of endoplasmic reticulum stress-associated regulatory proteins including ATF4 and B-lineage mutation of the pro-apoptotic ATF4 target Chop prevented bone marrow plasma cell demise on P2RX4 inhibition. Thus, generating mature protective and pathogenic plasma cells requires P2RX4 signalling controlled by PANX3-regulated extracellular ATP release from bone marrow niche cells.


Assuntos
Trifosfato de Adenosina , Células da Medula Óssea , Plasmócitos , Animais , Camundongos , Trifosfato de Adenosina/metabolismo , Autoanticorpos/imunologia , Autoimunidade/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Linhagem da Célula , Conexinas/genética , Conexinas/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Mutação , Osteoblastos/metabolismo , Plasmócitos/citologia , Plasmócitos/imunologia , Plasmócitos/metabolismo , Receptores Purinérgicos P2X4/metabolismo , Transdução de Sinais
3.
Sci Adv ; 9(46): eadg8126, 2023 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-37967174

RESUMO

Thymic epithelial cells (TEC) control T cell development and play essential roles in establishing self-tolerance. By using Foxn1-Cre-driven ablation of Klf6 gene in TEC, we identified Klf6 as a critical factor in TEC development. Klf6 deficiency resulted in a hypoplastic thymus-evident from fetal stages into adulthood-in which a dramatic increase in the frequency of apoptotic TEC was observed. Among cortical TEC (cTEC), a previously unreported cTEC population expressing the transcription factor Sox10 was relatively expanded. Within medullary TEC (mTEC), mTEC I and Tuft-like mTEC IV were disproportionately decreased. Klf6 deficiency altered chromatin accessibility and affected TEC chromatin configuration. Consistent with these defects, naïve conventional T cells and invariant natural killer T cells were reduced in the spleen. Late stages of T cell receptor-dependent selection of thymocytes were affected, and mice exhibited autoimmunity. Thus, Klf6 has a prosurvival role and affects the development of specific TEC subsets contributing to thymic function.


Assuntos
Regulação da Expressão Gênica , Timócitos , Animais , Camundongos , Diferenciação Celular/genética , Cromatina/metabolismo , Células Epiteliais/metabolismo , Camundongos Endogâmicos C57BL , Timócitos/metabolismo , Timo/metabolismo
4.
Front Immunol ; 11: 470, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32265924

RESUMO

The transcription factor TCF-1 (encoded by Tcf7) plays critical roles in several lineages of hematopoietic cells. In this study, we examined the molecular basis for Tcf7 regulation in T cells, innate lymphoid cells, and migratory conventional dendritic cells that we find express Tcf7. We identified a 1 kb regulatory element crucial for the initiation of Tcf7 expression in T cells and innate lymphoid cells, but dispensable for Tcf7 expression in Tcf7-expressing dendritic cells. Within this region, we identified a Notch binding site important for the initiation of Tcf7 expression in T cells but not in innate lymphoid cells. Our work establishes that the same regulatory element is used by distinct transcriptional controllers to initiate Tcf7 expression in T cells and ILCs.


Assuntos
Fator 1-alfa Nuclear de Hepatócito/metabolismo , Linfócitos/imunologia , Elementos Reguladores de Transcrição/genética , Linfócitos T/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/genética , Imunidade Inata , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Nat Commun ; 10(1): 5498, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31792212

RESUMO

Interactions between thymic epithelial cells (TEC) and developing thymocytes are essential for T cell development, but molecular insights on TEC and thymus homeostasis are still lacking. Here we identify distinct transcriptional programs of TEC that account for their age-specific properties, including proliferation rates, engraftability and function. Further analyses identify Myc as a regulator of fetal thymus development to support the rapid increase of thymus size during fetal life. Enforced Myc expression in TEC induces the prolonged maintenance of a fetal-specific transcriptional program, which in turn extends the growth phase of the thymus and enhances thymic output; meanwhile, inducible expression of Myc in adult TEC similarly promotes thymic growth. Mechanistically, this Myc function is associated with enhanced ribosomal biogenesis in TEC. Our study thus identifies age-specific transcriptional programs in TEC, and establishes that Myc controls thymus size.


Assuntos
Células Epiteliais/metabolismo , Proteína Oncogênica p55(v-myc)/metabolismo , Timo/embriologia , Transcrição Gênica , Animais , Células Epiteliais/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteína Oncogênica p55(v-myc)/genética , Tamanho do Órgão , Organogênese , Timo/metabolismo
6.
J Immunol ; 203(3): 686-695, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31243087

RESUMO

The thymus is critical for the establishment of the adaptive immune system and the development of a diverse T cell repertoire. T cell development depends upon cell-cell interactions with epithelial cells in the thymus. The thymus is composed of two different types of epithelial cells: cortical and medullary epithelial cells. Both of these express and critically depend on the transcription factor Foxn1 Foxn1 is also expressed in the hair follicle, and disruption of Foxn1 function in mice results in severe thymic developmental defects and the hairless (nude) phenotype. Despite its importance, little is known about the direct regulation of Foxn1 expression. In this study, we identify a cis-regulatory element (RE) critical for expression of Foxn1 in mouse thymic epithelial cells but dispensable for expression in hair follicles. Analysis of chromatin accessibility, histone modifications, and sequence conservation identified regions within the first intron of Foxn1 that possessed the characteristics of REs. Systematic knockout of candidate regions lead us to identify a 1.6 kb region that, when deleted, results in a near total disruption of thymus development. Interestingly, Foxn1 expression and function in the hair follicle were unaffected. RNA fluorescent in situ hybridization showed a near complete loss of Foxn1 mRNA expression in the embryonic thymic bud. Our studies have identified a genomic RE with thymic-specific control of Foxn1 gene expression.


Assuntos
Células Epiteliais/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Elementos Reguladores de Transcrição/genética , Linfócitos T/imunologia , Timo/metabolismo , Animais , Fatores de Transcrição Forkhead/biossíntese , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Folículo Piloso/metabolismo , Camundongos , Camundongos Knockout , Camundongos Nus , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Linfócitos T/citologia , Timo/citologia
7.
F1000Res ; 7: 318, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29707204

RESUMO

Background: We previously identified the transcriptional regulator Zbtb32 as a factor that can promote T cell tolerance in the Non-Obese Diabetic (NOD) mouse, a model of Type 1 diabetes. Antigen targeted to DCIR2 + dendritic cells (DCs) in vivo inhibited both diabetes and effector T cell expansion in NOD mice. Furthermore, Zbtb32 was preferentially induced in autoreactive CD4 T cells stimulated by these tolerogenic DCIR2 + DCs, and overexpression of Zbtb32 in islet-specific T cells inhibited the diabetes development by limiting T cell proliferation and cytokine production. Methods: To further understand the role of Zbtb32 in T cell tolerance induction, we have now used CRISPR to target the Zbtb32 gene for deletion directly in NOD mice and characterized the mutant mice. We hypothesized that the systemic loss of Zbtb32 in NOD mice would lead to increased T cell activation and increased diabetes pathogenesis. Results: Although NOD.Zbtb32 -/- male NOD mice showed a trend towards increased diabetes incidence compared to littermate controls, the difference was not significant. Furthermore, no significant alteration in lymphocyte number or function was observed. Importantly, in vitro stimulation of lymphocytes from NOD.Zbtb32 -/- mice did not produce the expected hypersensitive phenotype observed in other genetic strains, potentially due to compensation by homologous genes. Conclusions: The loss of Zbtb32 in the NOD background does not result in the expected T cell activation phenotype.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Diabetes Mellitus Experimental/epidemiologia , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Proteínas Repressoras/fisiologia , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Citocinas/metabolismo , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Feminino , Incidência , Masculino , Camundongos , Camundongos Endogâmicos NOD , Proteínas Repressoras/antagonistas & inibidores
8.
JCI Insight ; 3(3)2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29415894

RESUMO

Type I IFN (IFN-I) dysregulation contributes to type 1 diabetes (T1D) development, and although increased IFN-I signals are pathogenic at the initiation of autoimmune diabetes, IFN-I dysregulation at later pathogenic stages more relevant for therapeutic intervention is not well understood. We discovered that 5 key antigen-presenting cell subsets from adult prediabetic NOD mice have reduced responsiveness to IFN-I that is dominated by a decrease in the tonic-sensitive subset of IFN-I response genes. Blockade of IFNAR1 in prediabetic NOD mice accelerated diabetes and increased Th1 responses. Therefore, IFN-I responses shift from pathogenic to protective as autoimmunity progresses, consistent with chronic IFN-I exposure. In contrast, IL-1-associated inflammatory pathways were elevated in prediabetic mice. These changes correlated with human T1D onset-associated gene expression. Prostaglandin E2 (PGE2) and prostaglandin receptor 4 (PTGER4), a receptor for PGE2 that mediates both inflammatory and regulatory eicosanoid signaling, were higher in NOD mice and drive innate immune dysregulation. Treating prediabetic NOD mice with a PTGER4 antagonist restored IFNAR signaling, decreased IL-1 signaling, and decreased infiltration of leukocytes into the islets. Therefore, innate cytokine alterations contribute to both T1D-associated inflammation and autoimmune pathogenesis. Modulating innate immune balance via signals such as PTGER4 may contribute to treatments for autoimmunity.


Assuntos
Autoimunidade/efeitos dos fármacos , Diabetes Mellitus Tipo 1/imunologia , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Compostos de Sulfonilureia/administração & dosagem , Células Th1/imunologia , Administração Oral , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Dinoprostona/imunologia , Dinoprostona/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-1/imunologia , Interleucina-1/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/imunologia , Receptor de Interferon alfa e beta/metabolismo , Receptores de Prostaglandina E Subtipo EP4/imunologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/metabolismo
9.
J Autoimmun ; 90: 39-48, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29439835

RESUMO

Dendritic cell (DC)-mediated T cell tolerance deficiencies contribute to the pathogenesis of autoimmune diseases such as type 1 diabetes. Delivering self-antigen to dendritic-cell inhibitory receptor-2 (DCIR2)+ DCs can delay but not completely block diabetes development in NOD mice. These DCIR2-targeting antibodies induce tolerance via deletion and anergy, but do not increase islet-specific Tregs. Because low-dose IL-2 (LD-IL-2) administration can preferentially expand Tregs, we tested whether delivering islet-antigen to tolerogenic DCIR2+ DCs along with LD-IL-2 would boost islet-specific Tregs and further block autoimmunity. But, surprisingly, adding LD-IL-2 did not increase efficacy of DC-targeted antigen to inhibit diabetes. Here we show the effects of LD-IL-2, with or without antigen delivery to DCIR2+ DCs, on both polyclonal and autoreactive Treg and conventional T cells (Tconv). As expected, LD-IL-2 increased total Tregs, but autoreactive Tregs required both antigen and IL-2 stimulation for optimal expansion. Also, islet-specific Tregs had lower CD25 expression and IL-2 sensitivity, while islet-specific Tconv had higher CD25 expression, compared to polyclonal populations. LD-IL-2 increased activation and expansion of Tconv, and was more pronounced for autoreactive cells after treatment with IL-2 + islet-antigen. Therefore, LD-IL-2 therapy, especially when combined with antigen stimulation, may not optimally activate and expand antigen-specific Tregs in chronic autoimmune settings.


Assuntos
Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Interleucina-2/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Apresentação de Antígeno , Autoantígenos/imunologia , Autoimunidade , Células Cultivadas , Feminino , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Receptores de Superfície Celular/metabolismo
10.
J Exp Med ; 214(9): 2523-2533, 2017 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-28710273

RESUMO

Patients with hypomorphic mutations in STAT3 and patients with hypermorphic mutations in STAT1 share several clinical and cellular phenotypes suggesting overlapping pathophysiologic mechanisms. We, therefore, examined cytokine signaling and CD4+ T cell differentiation in these cohorts to characterize common pathways. As expected, differentiation of Th17 cells was impaired in both cohorts. We found that STAT1 was hyperphosphorylated in response to cytokine stimulation in both cohorts and that STAT1-dependent PD-L1 up-regulation-known to inhibit Th17 differentiation in mouse models-was markedly enhanced as well. Overexpression of SOCS3 strongly inhibited phosphorylation of STAT1 and PD-L1 up-regulation, suggesting that diminished SOCS3 expression may lead to the observed effects. Defects in Th17 differentiation could be partially overcome in vitro via PD-L1 inhibition and in a mouse model of STAT3 loss-of-function by crossing them with PD-1 knockout mice. PD-L1 may be a potential therapeutic target in several genetic diseases of immune deficiency affecting cytokine signaling.


Assuntos
Antígeno B7-H1/fisiologia , Diferenciação Celular/fisiologia , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT3/fisiologia , Células Th17/fisiologia , Adolescente , Adulto , Animais , Criança , Citocinas/fisiologia , Feminino , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/fisiopatologia , Interleucinas/fisiologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas/fisiologia , Regulação para Cima , Adulto Jovem
11.
PLoS Biol ; 14(6): e1002502, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27337557

RESUMO

[This corrects the article DOI: 10.1371/journal.pbio.1000518.].

12.
J Immunol ; 196(5): 2031-40, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26826238

RESUMO

Innate immune signals help break self-tolerance to initiate autoimmune diseases such as type 1 diabetes, but innate contributions to subsequent regulation of disease progression are less clear. Most studies have measured in vitro innate responses of GM-CSF dendritic cells (DCs) that are functionally distinct from conventional DCs (cDCs) and do not reflect in vivo DC subsets. To determine whether autoimmune NOD mice have alterations in type 1 IFN innate responsiveness, we compared cDCs from prediabetic NOD and control C57BL/6 (B6) mice stimulated in vivo with the TLR9 ligand CpG, a strong type 1 IFN inducer. In response to CpG, NOD mice produce more type 1 IFN and express higher levels of CD40, and NOD monocyte DCs make more TNF. However, the overall CpG-induced transcriptional response is muted in NOD cDCs. Of relevance the costimulatory proteins CD80/CD86, signals needed for regulatory T cell homeostasis, are upregulated less on NOD cDCs. Interestingly, NOD Rag1(-/-) mice also display a defect in CpG-induced CD86 upregulation compared with B6 Rag1(-/-), indicating this particular innate alteration precedes adaptive autoimmunity. The impaired response in NOD DCs is likely downstream of the IFN-α/ß receptor because DCs from NOD and B6 mice show similar CpG-induced CD86 levels when anti-IFN-α/ß receptor Ab is added. IFN-α-induced nuclear localization of activated STAT1 is markedly reduced in NOD CD11c(+) cells, consistent with lower type 1 IFN responsiveness. In conclusion, NOD DCs display altered innate responses characterized by enhanced type 1 IFN and activation of monocyte-derived DCs but diminished cDC type 1 IFN response.


Assuntos
Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Fator de Transcrição STAT1/imunologia , Tolerância a Antígenos Próprios/imunologia , Transporte Ativo do Núcleo Celular , Animais , Western Blotting , Linhagem da Célula , Núcleo Celular/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Microscopia Confocal , Monócitos/citologia , Monócitos/imunologia , Oligodesoxirribonucleotídeos/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT1/metabolismo , Receptor Toll-Like 9/agonistas
13.
J Biol Chem ; 290(36): 22076-84, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26224629

RESUMO

NF-κB essential modulator (NEMO) and cylindromatosis protein (CYLD) are intracellular proteins that regulate the NF-κB signaling pathway. Although mice with either CYLD deficiency or an alteration in the zinc finger domain of NEMO (K392R) are born healthy, we found that the combination of these two gene defects in double mutant (DM) mice is early embryonic lethal but can be rescued by the absence of TNF receptor 1 (TNFR1). Notably, NEMO was not recruited into the TNFR1 complex of DM cells, and consequently NF-κB induction by TNF was severely impaired and DM cells were sensitized to TNF-induced cell death. Interestingly, the TNF signaling defects can be fully rescued by reconstitution of DM cells with CYLD lacking ubiquitin hydrolase activity but not with CYLD mutated in TNF receptor-associated factor 2 (TRAF2) or NEMO binding sites. Therefore, our data demonstrate an unexpected non-catalytic function for CYLD as an adapter protein between TRAF2 and the NEMO zinc finger that is important for TNF-induced NF-κB signaling during embryogenesis.


Assuntos
Desenvolvimento Embrionário/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Ligação Proteica , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Dedos de Zinco/genética
15.
Diabetes ; 64(10): 3521-31, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26070317

RESUMO

During autoimmunity, the normal ability of dendritic cells (DCs) to induce T-cell tolerance is disrupted; therefore, autoimmune disease therapies based on cell types and molecular pathways that elicit tolerance in the steady state may not be effective. To determine which DC subsets induce tolerance in the context of chronic autoimmunity, we used chimeric antibodies specific for DC inhibitory receptor 2 (DCIR2) or DEC-205 to target self-antigen to CD11b(+) (cDC2) DCs and CD8(+) (cDC1) DCs, respectively, in autoimmune-prone nonobese diabetic (NOD) mice. Antigen presentation by DCIR2(+) DCs but not DEC-205(+) DCs elicited tolerogenic CD4(+) T-cell responses in NOD mice. ß-Cell antigen delivered to DCIR2(+) DCs delayed diabetes induction and induced increased T-cell apoptosis without interferon-γ (IFN-γ) or sustained expansion of autoreactive CD4(+) T cells. These divergent responses were preceded by differential gene expression in T cells early after in vivo stimulation. Zbtb32 was higher in T cells stimulated with DCIR2(+) DCs, and overexpression of Zbtb32 in T cells inhibited diabetes development, T-cell expansion, and IFN-γ production. Therefore, we have identified DCIR2(+) DCs as capable of inducing antigen-specific tolerance in the face of ongoing autoimmunity and have also identified Zbtb32 as a suppressive transcription factor that controls T cell-mediated autoimmunity.


Assuntos
Anticorpos , Antígenos CD/metabolismo , Autoimunidade/fisiologia , Linfócitos T CD4-Positivos/fisiologia , Células Dendríticas/fisiologia , Diabetes Mellitus/imunologia , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Repressoras/metabolismo , Animais , Antígenos CD/genética , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/genética , Antígenos CD40/metabolismo , Diabetes Mellitus/prevenção & controle , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica/imunologia , Lectinas Tipo C/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Antígenos de Histocompatibilidade Menor , Receptores de Superfície Celular/genética , Proteínas Repressoras/genética , Organismos Livres de Patógenos Específicos
16.
Proc Natl Acad Sci U S A ; 111(38): 13930-5, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25201955

RESUMO

Novel inhibitor of histone acetyltransferase repressor (NIR) is a transcriptional corepressor with inhibitor of histone acetyltransferase activity and is a potent suppressor of p53. Although NIR deficiency in mice leads to early embryonic lethality, lymphoid-restricted deletion resulted in the absence of double-positive CD4(+)CD8(+) thymocytes, whereas bone-marrow-derived B cells were arrested at the B220(+)CD19(-) pro-B-cell stage. V(D)J recombination was preserved in NIR-deficient DN3 double-negative thymocytes, suggesting that NIR does not affect p53 function in response to physiologic DNA breaks. Nevertheless, the combined deficiency of NIR and p53 provided rescue of DN3L double-negative thymocytes and their further differentiation to double- and single-positive thymocytes, whereas B cells in the marrow further developed to the B220(+)CD19(+) pro-B-cell stage. Our results show that NIR cooperate with p53 to impose checkpoint for the generation of mature B and T lymphocytes.


Assuntos
Diferenciação Celular/imunologia , Proteínas Repressoras/imunologia , Timócitos/imunologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular/genética , Quebras de DNA , Camundongos , Células Precursoras de Linfócitos B/citologia , Proteínas Repressoras/genética , Timócitos/citologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia
17.
Nat Commun ; 5: 4585, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25134987

RESUMO

CYLD is a tumour suppressor gene mutated in familial cylindromatosis, a genetic disorder leading to the development of skin appendage tumours. It encodes a deubiquitinating enzyme that removes Lys63- or linear-linked ubiquitin chains. CYLD was shown to regulate cell proliferation, cell survival and inflammatory responses, through various signalling pathways. Here we show that CYLD localizes at centrosomes and basal bodies via interaction with the centrosomal protein CAP350 and demonstrate that CYLD must be both at the centrosome and catalytically active to promote ciliogenesis independently of NF-κB. In transgenic mice engineered to mimic the smallest truncation found in cylindromatosis patients, CYLD interaction with CAP350 is lost disrupting CYLD centrosome localization, which results in cilia formation defects due to impairment of basal body migration and docking. These results point to an undiscovered regulation of ciliogenesis by Lys63 ubiquitination and provide new perspectives regarding CYLD function that should be considered in the context of cylindromatosis.


Assuntos
Corpos Basais/fisiologia , Comunicação Celular/fisiologia , Centrossomo/fisiologia , Cílios/fisiologia , Cisteína Endopeptidases/fisiologia , Células Epiteliais/fisiologia , Animais , Células Cultivadas , Cisteína Endopeptidases/genética , Proteínas do Citoesqueleto/fisiologia , Enzima Desubiquitinante CYLD , Células Epiteliais/citologia , Feminino , Humanos , Rim/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microtúbulos/fisiologia , NF-kappa B/fisiologia , Proteínas Nucleares/fisiologia , Retina/citologia , Transdução de Sinais/fisiologia
18.
Immunology ; 143(4): 640-50, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24954893

RESUMO

We have previously shown that interleukin-2 (IL-2) inhibits dendritic cell (DC) development from mouse bone marrow (BM) precursors stimulated with the ligand for FMS-like tyrosine kinase 3 receptor (Flt3L), and have provided evidence that this inhibition occurs at the monocyte DC precursor stage of DC development. Here, we explored the mechanism of IL-2-mediated inhibition of DC development. First, we showed that these in vitro cultures accurately model DCs that develop in vivo by comparing gene and protein expression of the three main Flt3L-induced DC subsets from the BM, CD11b(+) and CD24(+) conventional DCs (cDCs) and plasmacytoid DCs (pDCs) with their respective ex vivo spleen DC subsets (CD11b(+), CD8(+) and pDCs). Next, gene expression changes were quantified in Flt3L DC subsets that developed in the presence of IL-2. These changes included increased expression of Bcl2l11, which encodes the apoptosis-inducing protein Bim, and decreased expression of Flt3 (CD135), the receptor that initiates DC development. Interleukin-2 also significantly reduced Flt3 protein expression on all three Flt3L DC subsets, and attenuated Flt3L-induced STAT3 phosphorylation in DCs. Based on these data, we hypothesized that decreased Flt3 signalling may divert BM precursors down monocyte and macrophage lineages. Indeed, addition of IL-2 led to increases in Flt3(-) cells, including cKit(+) Ly6C(+) CD11b(-) populations consistent with the recently identified committed monocyte/macrophage progenitor. Therefore, IL-2 can inhibit DC development via decreased signalling through Flt3 and increased monocyte/macrophage development.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Interleucina-2/farmacologia , Macrófagos/metabolismo , Monócitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Citocinas/genética , Células Dendríticas/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT3/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo
19.
J Leukoc Biol ; 95(2): 325-36, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24082013

RESUMO

DCs are important mediators of peripheral tolerance for the prevention of autoimmunity. Chimeric αDEC-205 antibodies with attached antigens allow in vivo antigen-specific stimulation of T cells by CD8(+) DCs, resulting in tolerance in nonautoimmune mice. However, it is not clear whether DC-mediated tolerance induction occurs in the context of ongoing autoimmunity. We assessed the role of CD8(+) DCs in stimulation of autoreactive CD4(+) T cells in the NOD mouse model of type 1 diabetes. Targeting of antigen to CD8(+) DCs via αDEC-205 led to proliferation and expansion of ß-cell specific BDC2.5 T cells. These T cells also produced IL-2 and IFN-γ and did not up-regulate FoxP3, consistent with an activated rather than tolerant phenotype. Similarly, endogenous BDC peptide-reactive T cells, identified with I-A(g7) tetramers, did not become tolerant after antigen delivery via αDEC-205: no deletion or Treg induction was observed. We observed that CD8(+) DCs from NOD mice expressed higher surface levels of CD40 than CD8(+) DCs from C57BL/6 mice. Blockade of CD40-CD40L interactions reduced the number of BDC2.5 T cells remaining in mice, 10 days after antigen targeting to CD8 DCs, and blocked IFN-γ production by BDC2.5 T cells. These data indicate that the ability of autoreactive CD4(+) T cells to undergo tolerance mediated by CD8(+) DCs is defective in NOD mice and that blocking CD40-CD40L interactions can restore tolerance induction.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Ligante de CD40/antagonistas & inibidores , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Tolerância Imunológica/imunologia , Animais , Anticorpos/farmacologia , Antígenos/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Antígenos CD40/metabolismo , Ligante de CD40/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Fatores de Transcrição Forkhead/metabolismo , Tolerância Imunológica/efeitos dos fármacos , Interferon gama/metabolismo , Ligantes , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos NOD , Peptídeos/imunologia , Fenótipo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th1/efeitos dos fármacos , Células Th1/metabolismo , Receptores Toll-Like/metabolismo
20.
Proc Natl Acad Sci U S A ; 110(13): 5127-32, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23493554

RESUMO

Mutations in the TNF family of proteins have been associated with inherited forms of immune deficiency. Using an array-based sequencing assay, we identified an autosomal-dominant deficiency in TNF-like weak inducer of apoptosis (TWEAK; TNFSF12) in a kindred with recurrent infection and impaired antibody responses to protein and polysaccharide vaccines. This mutation occurs in the sixth exon of TWEAK and results in the amino acid substitution R145C within the conserved TNF-homology domain of the full-length protein. TWEAK mutant protein formed high molecular weight aggregates under nonreducing conditions, suggesting an increased propensity for intermolecular interactions. As a result, mutant TWEAK associated with B-cell-activating factor (BAFF) protein and down-regulated the BAFF-mediated activation of the noncanonical NF-κB pathway through inhibition of p100 processing to p52, resulting in inhibition of BAFF-dependent B-cell survival and proliferation. As BAFF mediates T-cell-independent isotype switching and B-cell survival, our data implicate TWEAK as a disease-susceptibility gene for a humoral immunodeficiency.


Assuntos
Linfócitos B/imunologia , Doenças Genéticas Inatas/imunologia , Predisposição Genética para Doença , Síndromes de Imunodeficiência/imunologia , Mutação de Sentido Incorreto , Fatores de Necrose Tumoral/imunologia , Adulto , Substituição de Aminoácidos , Fator Ativador de Células B/genética , Fator Ativador de Células B/imunologia , Linfócitos B/patologia , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Criança , Pré-Escolar , Citocina TWEAK , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Feminino , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/patologia , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/patologia , Masculino , Subunidade p52 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/imunologia , Fatores de Necrose Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA