Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transl Res ; 255: 37-49, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36400308

RESUMO

Dendritic cells (DCs) are key regulators of the adaptive immune response. Tolerogenic dendritic cells play a crucial role in inducing and maintaining immune tolerance in autoimmune diseases such as type 1 diabetes in humans as well as in the NOD mouse model. We previously reported that bone marrow-derived DCs (BM.DCs) from NOD mice, generated with a low dose of GM-CSF (GM/DCs), induce Treg differentiation and are able to protect NOD mice from diabetes. We had also found that the p38 MAPK/C/EBPß axis is involved in regulating the phenotype, as well as the production of IL-10 and IL-12p70, by tolerogenic GM/DCs. Here, we report that the inhibition of the PI3K signaling switched the cytokine profile of GM/DCs toward Th17-promoting cytokines without affecting their phenotype. PI3K inhibition abrogated the production of IL-10 by GM/DCs, whereas it enhanced their production of IL-23 and TGFß. Inhibition of PI3K signaling in tolerogenic GM/DCs also induced naive CD4+ T cells differentiation toward Th17 cells. Mechanistically, PI3K inhibition increased the DNA-binding activity of C/EBPß through a GSK3-dependent pathway, which is important to maintain the semimature phenotype of tolerogenic GM/DCs. Furthermore, analysis of C/EBPß-/- GM/DCs demonstrated that C/EBPß is required for IL-23 production. Of physiological relevance, the level of protection from diabetes following transfusion of GM/DCs into young NOD mice was significantly reduced when NOD mice were transfused with GM/DCs pretreated with a PI3K inhibitor. Our data suggest that PI3K/C/EBPß signaling is important in controlling tolerogenic function of GM/DCs by limiting their Th17-promoting cytokines.


Assuntos
Diabetes Mellitus , Interleucina-10 , Humanos , Camundongos , Animais , Camundongos Endogâmicos NOD , Fosfatidilinositol 3-Quinases/metabolismo , Células Th17/metabolismo , Medula Óssea , Quinase 3 da Glicogênio Sintase/metabolismo , Linfócitos T Reguladores , Diferenciação Celular , Citocinas/metabolismo , Tolerância Imunológica , Células Dendríticas/metabolismo , Interleucina-23/metabolismo , Diabetes Mellitus/metabolismo
2.
Cells ; 7(12)2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30544623

RESUMO

Dendritic cells (DCs) play a major role in innate and adaptive immunity and self-immune tolerance. Immunogenic versus tolerogenic DC functions are dictated by their levels of costimulatory molecules and their cytokine expression profile. The transcription factor C/EBPß regulates the expression of several inflammatory genes in many cell types including macrophages. However, little is known regarding the role of C/EBPß in tolerogenic versus immunogenic DCs functions. We have previously reported that bone marrow-derived DCs generated with GM-CSF (GM/DCs) acquire the signature of semi-mature tolerogenic IL-10-producing DCs as opposed to immunogenic DCs generated with GM-CSF and IL-4 (IL-4/DCs). Here, we show that tolerogenic GM/DCs exhibit higher levels of phosphorylation and enhanced DNA binding activity of C/EBPß and CREB than immunogenic IL-4/DCs. We also show that the p38 MAPK/CREB axis and GSK3 play an important role in regulating C/EBPß phosphorylation and DNA binding activity. Inhibition of p38 MAPK in GM/DCs resulted in a drastic decrease of C/EBPß and CREB DNA binding activities, a reduction of their IL-10 production and an increase of their IL-12p70 production, a characteristic of immunogenic IL-4/DCs. We also present evidence that GSK3 inhibition in GM/DCs reduced C/EBPß DNA binding activity and increased expression of costimulatory molecules in GM/DCs and their production of IL-10. Analysis of GM/DCs of C/EBPß-/- mice showed that C/EBPß was essential to maintain the semimature phenotype and the production of IL-10 as well as low CD4⁺ T cell proliferation. Our results highlight the importance of the p38MAPK-C/EBPß pathway in regulating phenotype and function of tolerogenic GM/DCs.

3.
J Autoimmun ; 76: 63-74, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27634616

RESUMO

Defects in dendritic cells (DCs) development and function lead to autoimmune disorders. Autoimmune diabetes in humans and NOD mice results from a breakdown of self-tolerance, ending in T cell-mediated ß-cell destruction. DCs dysfunction in NOD mice results in part from a defect in the JAK-STAT5 signaling pathway associated with the idd4 susceptibility locus. The involvement of Stat5b in DCs tolerogenic functions remains unknown. We have generated transgenic mice (NOD.CD11cStat5b-CA) expressing a constitutively active form of the Stat5b gene (Stat5b-CA) under control of CD11c promoter. All NOD.CD11cStat5b-CA mice were protected against diabetes. Protection was associated with an increased in the pool and suppressive function of Tregs, a promotion of Th2 and Tc2 immune response and a decreased percentage of CD8+ T cells. Splenic DCs of NOD.CD11cStat5b-CA mice acquired a mature phenotype, promoted and induced better conversion of CD4+CD25-Foxp3- T cells into Tregs (CD4+CD25+Foxp3+ T cells) than DCs of NOD mice. Stat5b-CA.DC-educated CD4+CD25- T cells delayed diabetes onset whereas Stat5b-CA.DC-educated Tregs blocked ongoing diabetes in 8-10 weeks old NOD recipient mice. Importantly, injection of Stat5b.CA.DC to 8-10-week old NOD mice halted diabetes progression and educated their splenocytes to loose their diabetogenic potential when transferred to NOD.SCID mice. Our work is the first to report that an active form of Stat5b restored DCs tolerogenic functions that re-educated Tregs to re-establish and to sustain long-term protective immune response against diabetes in NOD mice.


Assuntos
Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Fator de Transcrição STAT5/metabolismo , Tolerância a Antígenos Próprios/imunologia , Transdução de Sinais , Animais , Autoantígenos/imunologia , Autoimunidade , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Progressão da Doença , Imunofenotipagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Fenótipo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
4.
Cell Immunol ; 272(2): 259-68, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22070873

RESUMO

Tolerogenic dendritic cells represent a promising immunotherapy in autoimmunity. However, the molecular mechanisms that drive tolerogenic DCs functions are not well understood. We used GM-CSF or GM-CSF+IL-4 to generate tolerogenic (GM/DCs) and immunogenic (IL-4/DCs) BMDCs from NOD mice, respectively. GM/DCs were resistant to maturation, produced large amounts of IL-10 but not IL-12p70. GM/DCs displayed a reduced capacity to activate diabetogenic CD8(+) T-cells and were efficient to induce Tregs expansion and conversion. LPS stimulation triggered ERK1/2 activation that was sustained in GM/DCs but not in IL-4/DCs. ERK1/2 and AP-1 were involved in IL-10 production in GM/DCs but not in their resistance to maturation. Supershift analysis showed that NF-κB DNA binding complex contains p52 and p65 in GM/DCs, whereas it contains p52, p65 and RelB in IL-4/DCs. ChIP experiments revealed that p65 was recruited to IL-10 promoter following LPS stimulation of GM/DCs whereas its binding to IL-12p35 promoter was abolished. Our results suggest that immunoregulatory functions of GM/DCs are differentially regulated by ERK1/2, AP-1 and NF-κB pathways.


Assuntos
Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , NF-kappa B/imunologia , Fator de Transcrição AP-1/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Processos de Crescimento Celular/imunologia , Imunoprecipitação da Cromatina/métodos , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-12/imunologia , Subunidade p35 da Interleucina-12/genética , Subunidade p35 da Interleucina-12/imunologia , Interleucina-4/imunologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Subunidade p52 de NF-kappa B/imunologia , Fenótipo , Regiões Promotoras Genéticas , Linfócitos T Reguladores/imunologia , Fator de Transcrição RelA/imunologia , Fator de Transcrição RelB/imunologia
5.
BMC Musculoskelet Disord ; 12: 185, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21843349

RESUMO

BACKGROUND: Skeletal muscle atrophy is a serious concern for the rehabilitation of patients afflicted by prolonged limb restriction. This debilitating condition is associated with a marked activation of NFκB activity. The ubiquitin-proteasome pathway degrades the NFκB inhibitor IκBα, enabling NFκB to translocate to the nucleus and bind to the target genes that promote muscle atrophy. Although several studies showed that proteasome inhibitors are efficient to reduce atrophy, no studies have demonstrated the ability of these inhibitors to preserve muscle function under catabolic condition. METHODS: We recently developed a new hindlimb immobilization procedure that induces significant skeletal muscle atrophy and used it to show that an inflammatory process characterized by the up-regulation of TNFα, a known activator of the canonical NFκB pathway, is associated with the atrophy. Here, we used this model to investigate the effect of in vivo proteasome inhibition on the muscle integrity by histological approach. TNFα, IL-1, IL-6, MuRF-1 and Atrogin/MAFbx mRNA level were determined by qPCR. Also, a functional measurement of locomotors activity was performed to determine if the treatment can shorten the rehabilitation period following immobilization. RESULTS: In the present study, we showed that the proteasome inhibitor MG132 significantly inhibited IκBα degradation thus preventing NFκB activation in vitro. MG132 preserved muscle and myofiber cross-sectional area by downregulating the muscle-specific ubiquitin ligases atrogin-1/MAFbx and MuRF-1 mRNA in vivo. This effect resulted in a diminished rehabilitation period. CONCLUSION: These finding demonstrate that proteasome inhibitors show potential for the development of pharmacological therapies to prevent muscle atrophy and thus favor muscle rehabilitation.


Assuntos
Inibidores de Cisteína Proteinase/farmacologia , Elevação dos Membros Posteriores/efeitos adversos , Leupeptinas/farmacologia , Atrofia Muscular/tratamento farmacológico , Inibidores de Proteassoma , Animais , Linhagem Celular , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Atrofia Muscular/reabilitação , Complexo de Endopeptidases do Proteassoma/fisiologia
6.
Clin Dev Immunol ; 2011: 374859, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21647406

RESUMO

Dendritic cells (DCs) contribute to islet inflammation and its progression to diabetes in NOD mouse model and human. DCs play a crucial role in the presentation of autoantigen and activation of diabetogenic T cells, and IRF4 and IRF8 are crucial genes involved in the development of DCs. We have therefore investigated the expression of these genes in splenic DCs during diabetes progression in NOD mice. We found that IRF4 expression was upregulated in splenocytes and in splenic CD11c(+) DCs of NOD mice as compared to BALB/c mice. In contrast, IRF8 gene expression was higher in splenocytes of NOD mice whereas its expression was similar in splenic CD11c(+) DCs of NOD and BALB/c mice. Importantly, levels of IRF4 and IRF8 expression were lower in tolerogenic bone marrow derived DCs (BMDCs) generated with GM-CSF as compared to immunogenic BMDCs generated with GM-CSF and IL-4. Analysis of splenic DCs subsets indicated that high expression of IRF4 was associated with increased levels of CD4(+)CD8α(-)IRF4(+)CD11c(+) DCs but not CD4(-)CD8α(+)IRF8(+)CD11c(+) DCs in NOD mice. Our results showed that IRF4 expression was up-regulated in NOD mice and correlated with the increased levels of CD4(+)CD8α(-) DCs, suggesting that IRF4 may be involved in abnormal DC functions in type 1 diabetes in NOD mice.


Assuntos
Regulação da Expressão Gênica , Fatores Reguladores de Interferon/genética , Animais , Células da Medula Óssea/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fatores Reguladores de Interferon/metabolismo , Interleucina-4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Regulação para Cima
7.
Cell Immunol ; 265(1): 31-6, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20637454

RESUMO

We have reported that GM-CSF treatment of NOD mice suppressed diabetes by increasing the number of tolerogenic dendritic cells (tDCs) and Tregs in the periphery. Here, we have investigated whether GM-CSF acted on NOD bone marrow DCs precursors to skew their differentiation to tDCs. DCs were generated from the bone marrow of GM-CSF-treated (GM.BMDCs) and PBS-treated (PBS.BMDCs) NOD mice and were assessed for their ability to acquire tolerogenic properties. Upon LPS stimulation, GM.BMDCs became fully mature, expressed high levels of PD-L1 and produced more IL-10 and less IL-12p70 and IFN-gamma than PBS.BMDCs. In addition, LPS-stimulated GM.BMDCs possessed a reduced capacity to activate diabetogenic CD8(+) T cells in a PD-1/PD-L1-dependent manner. A single injection of LPS-stimulated GM.BMDCs in NOD mice resulted in long-term protection from diabetes, in contrast to LPS-stimulated PBS.BMDCs. Our results showed that GM-CSF-treatment acted on bone marrow precursors to skew their differentiation into tDCs that protected NOD mice against diabetes.


Assuntos
Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Diabetes Mellitus/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Tolerância Imunológica/imunologia , Transferência Adotiva , Animais , Antígeno B7-1/imunologia , Antígeno B7-H1 , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Diabetes Mellitus/prevenção & controle , Feminino , Citometria de Fluxo , Histocitoquímica , Interferon gama/imunologia , Interleucina-10/imunologia , Interleucina-12/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Peptídeos/imunologia , Organismos Livres de Patógenos Específicos
8.
J Immunol ; 182(1): 563-71, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19109189

RESUMO

A growing number of neutrophil-derived cytokines have proven to be crucial to various inflammatory and immune processes in vivo. Whereas C/EBP (CCAAT/enhancer-binding protein) transcription factors are important for neutrophil differentiation from myeloid precursors, we report herein that they also regulate cytokine production in mature neutrophils. All known C/EBP proteins but C/EBPgamma are expressed in neutrophils; most isoforms localize to the nucleus, except for C/EBPalpha, which is cytoplasmic. Neutrophil stimulation does not alter the overall levels, cellular distribution, or turnover of C/EBP proteins; it also does not further induce the constitutive DNA-binding activity detected in nuclear extracts, consisting of C/EBPbeta and C/EBPepsilon. However, nuclear C/EBPbeta is rapidly phosphorylated upon cell stimulation, suggesting that it can activate cytokine promoters. Indeed, the transactivation of an IL-8 promoter-luciferase construct in a human neutrophil-like cell line was impaired when its C/EBP or NF-kappaB sites were mutated. Overexpression of a C/EBP repressor also impeded IL-8 promoter transactivation, as well as the generation of IL-8, Mip-1alpha, and Mip-1beta in this cellular model, whereas TNF-alpha generation was mostly unaffected. Finally, overexpression of a C/EBPbeta mutant (T235A) as well as chromatin immunoprecipitation assays unveiled an important role for this residue in cytokine induction. This is the first demonstration that C/EBP factors are important regulators of cytokine expression in human neutrophils.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Neutrófilos/imunologia , Neutrófilos/patologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Células CHO , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Cromatina/metabolismo , Cricetinae , Cricetulus , Regulação da Expressão Gênica/imunologia , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Neutrófilos/metabolismo , Fosforilação , Regiões Promotoras Genéticas/imunologia , Ligação Proteica/imunologia , Ativação Transcricional/imunologia
9.
Crit Care Med ; 36(11): 3054-61, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18824919

RESUMO

OBJECTIVE: Sepsis-induced organ dysfunctions remain prevalent and account for >50% of intensive care unit admissions for acute renal failure with a mortality rate nearing 75%. In addition to the fact that the mechanisms underlying the pathophysiology of sepsis-related acute renal failure are unclear, the impact on septic-induced acute renal failure of either norepinephrine, a gold-standard vasopressor, and arginine vasopressin, a candidate alternative, are not well understood. DESIGN: Randomized and controlled in vivo study. SETTING: Research laboratory and animal facilities. SUBJECTS: Adult rats treated with endotoxin (lipopolysaccharide) and/or vasopressors. INTERVENTIONS: Rats were intraperitoneally injected with lipopolysaccharide (12 mg/kg) or saline and then infused with either saline, 0.375 microg/microL arginine vasopressin, or 32.5 microg/microL norepinephrine for 18 hrs. These vasopressor rates yielded respective targeted blood levels observed in human septic shock. MEASUREMENTS AND MAIN RESULTS: Renal function, including glomerular filtration rate and fraction, renal blood flow, aquaporin-2, and arginine vasopressin-2 (V2 receptor) networking, water and salt handling, and urinary protein excretion, were evaluated. After lipopolysaccharide challenge arginine vasopressin infusion: 1) impaired creatinine clearance without affecting renal blood flow, glomerular filtration rate, and fraction but reduced free-water clearance, both of which being partially restored by the V2 receptor antagonist SR-121463B; 2) decreased the recognized ability of arginine vasopressin alone to recruit aquaporin-2 to the apical membrane increase its mRNA expression and urinary release; 3) increased urinary protein content but decreased specific kidney injury molecule-1, and Clara cell protein-16 release (p < 0.05 vs. lipopolysaccharide alone). Conversely, norepinephrine infusion did not add to lipopolysaccharide-induced alteration of urine biochemistry, except for improved creatinine clearance and increased microalbuminuria. CONCLUSION: In this endotoxic model, dose-targeted arginine vasopressin infusion increased lipopolysaccharide-induced renal dysfunction without affecting renal blood flow and glomerular function, but with particular disruption of aquaporin-2/V2 receptor networking, consecutive decreased salt and water handling ability. This is in clear contrast with norepinephrine infusion and suggests specific arginine vasopressin-induced "tubular epithelial dysfunction."


Assuntos
Injúria Renal Aguda/fisiopatologia , Aquaporina 2/análise , Endotoxinas/farmacologia , Rim/química , Lipopolissacarídeos/farmacologia , Receptores de Vasopressinas/análise , Vasoconstritores/farmacologia , Injúria Renal Aguda/etiologia , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos , Arginina Vasopressina/farmacologia , Creatinina/metabolismo , Epinefrina/farmacologia , Taxa de Filtração Glomerular , Injeções Intraperitoneais , Masculino , Morfolinas/farmacologia , Proteinúria/urina , RNA Mensageiro/análise , Distribuição Aleatória , Ratos , Ratos Wistar , Circulação Renal/fisiologia , Sepse/complicações , Compostos de Espiro/farmacologia , Uteroglobina/análise
10.
Diabetes ; 57(8): 2107-17, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18477807

RESUMO

OBJECTIVE: Autoimmune diabetes in the nonobese diabetic (NOD) mouse model results from a breakdown of T-cell tolerance caused by impaired tolerogenic dendritic cell development and regulatory T-cell (Treg) differentiation. Re-establishment of the Treg pool has been shown to confer T-cell tolerance and protection against diabetes. Here, we have investigated whether murine thymic stromal lymphopoietin (TSLP) re-established tolerogenic function of dendritic cells and induced differentiation and/or expansion of Tregs in NOD mice and protection against diabetes. RESEARCH DESIGN AND METHODS: We examined the phenotype of TSLP-conditioned bone marrow dendritic cells (TSLP-DCs) of NOD mice and their functions to induce noninflammatory Th2 response and differentiation of Tregs. The functional relevance of TSLP and TSLP-DCs to development of diabetes was also tested. RESULTS: Our results showed that bone marrow dendritic cells of NOD mice cultured in the presence of TSLP acquired signatures of tolerogenic dendritic cells, such as an absence of production of pro-inflammatory cytokines and a decreased expression of dendritic cell costimulatory molecules (CD80, CD86, and major histocompatibility complex class II) compared with LPS-treated dendritic cells. Furthermore, TSLP-DCs promoted noninflammatory Th2 response and induced the conversion of naïve T-cells into functional CD4(+)CD25(+)Foxp3(+) Tregs. We further showed that subcutaneous injections of TSLP for 6 days or a single intravenous injection of TSLP-DCs protected NOD mice against diabetes. CONCLUSIONS: Our study demonstrates that TSLP re-established a tolerogenic immune response in NOD mice and protects from diabetes, suggesting that TSLP may have a therapeutic potential for the treatment of type 1 diabetes.


Assuntos
Citocinas/farmacologia , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Antígeno B7-2/metabolismo , Antígenos CD8/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/transplante , Diabetes Mellitus Tipo 1/terapia , Feminino , Antígenos H-2/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfopoietina do Estroma do Timo
11.
J Immunol ; 179(6): 3638-47, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17785799

RESUMO

Autoimmune diabetes results from a breakdown of self-tolerance that leads to T cell-mediated beta-cell destruction. Abnormal maturation and other defects of dendritic cells (DCs) have been associated with the development of diabetes. Evidence is accumulating that self-tolerance can be restored and maintained by semimature DCs induced by GM-CSF. We have investigated whether GM-CSF is a valuable strategy to induce semimature DCs, thereby restoring and sustaining tolerance in NOD mice. We found that treatment of prediabetic NOD mice with GM-CSF provided protection against diabetes. The protection was associated with a marked increase in the number of tolerogenic immature splenic DCs and in the number of Foxp3+CD4+CD25+ regulatory T cells (Tregs). Activated DCs from GM-CSF-protected mice expressed lower levels of MHC class II and CD80/CD86 molecules, produced more IL-10 and were less effective in stimulating diabetogenic CD8+ T cells than DCs of PBS-treated NOD mice. Adoptive transfer experiments showed that splenocytes of GM-CSF-protected mice did not transfer diabetes into NOD.SCID recipients. Depletion of CD11c+ DCs before transfer released diabetogenic T cells from the suppressive effect of CD4+CD25+ Tregs, thereby promoting the development of diabetes. These results indicated that semimature DCs were required for the sustained suppressive function of CD4+CD25+ Tregs that were responsible for maintaining tolerance of diabetogenic T cells in NOD mice.


Assuntos
Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Tolerância a Antígenos Próprios , Linfócitos T Reguladores/imunologia , Animais , Apresentação de Antígeno/imunologia , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/patologia , Feminino , Inibidores do Crescimento/administração & dosagem , Imunofenotipagem , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Interleucina-10/biossíntese , Interleucina-10/fisiologia , Lipopolissacarídeos/administração & dosagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Estado Pré-Diabético/imunologia , Linfócitos T Reguladores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...