Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Leukoc Biol ; 113(1): 41-57, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36822162

RESUMO

Systemic lupus erythematosus development is influenced by both sex and the gut microbiota. Metabolite production is a major mechanism by which the gut microbiota influences the immune system, and we have previously found differences in the fecal metabolomic profiles of lupus-prone female and lupus-resistant male BWF1 mice. Here we determine how sex and microbiota metabolite production may interact to affect lupus. Transcriptomic analysis of female and male splenocytes showed genes that promote phagocytosis were upregulated in BWF1 male mice. Because patients with systemic lupus erythematosus exhibit defects in macrophage-mediated phagocytosis of apoptotic cells (efferocytosis), we compared splenic macrophage efferocytosis in vitro between female and male BWF1 mice. Macrophage efferocytosis was deficient in female compared to male BWF1 mice but could be restored by feeding male microbiota. Further transcriptomic analysis of the genes upregulated in male BWF1 mice revealed enrichment of genes stimulated by PPARγ and LXR signaling. Our previous fecal metabolomics analyses identified metabolites in male BWF1 mice that can activate PPARγ and LXR signaling and identified one in particular, phytanic acid, that is a very potent agonist. We show here that treatment of female BWF1 splenic macrophages with phytanic acid restores efferocytic activity via activation of the PPARγ and LXR signaling pathways. Furthermore, we found phytanic acid may restore female BWF1 macrophage efferocytosis through upregulation of the proefferocytic gene CD36. Taken together, our data indicate that metabolites produced by BWF1 male microbiota can enhance macrophage efferocytosis and, through this mechanism, could potentially influence lupus progression.


Assuntos
Lúpus Eritematoso Sistêmico , Microbiota , Camundongos , Masculino , Feminino , Animais , PPAR gama , Ácido Fitânico , Camundongos Endogâmicos NZB , Macrófagos , Fagocitose , Transdução de Sinais
2.
Immunol Cell Biol ; 98(8): 650-666, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32392367

RESUMO

The metabolically dynamic nature of healthy adipose places this tissue under regular inflammatory stress. A network of adipose-resident anti-inflammatory immune cells modulates and resolves this endogenous inflammation. Previous work in our laboratory identified a CD11b+ Gr1+ subset of these immunosuppressive adipose stromal cells in healthy mice. Myeloid-derived suppressor cells (MDSCs), typically associated with cancer and chronic inflammation, have a similar surface marker phenotype and accumulate in adipose of high-fat diet-fed mice. Given the routine inflammatory stresses on healthy adipose and the suppressive nature of the tissue-resident immune cells, we hypothesized that these CD11b+ Gr1+ cells were a genuine population of MDSCs involved in regulating tissue homeostasis. Flow cytometric analysis of these cells found that they were CD11b+ CD301- Ly6C+ Ly6G+/- and did not express traditional macrophage markers. Moreover, in vitro functional assays demonstrated that these cells suppressed αCD3/αCD28-induced T-cell proliferation, solidifying their identity as bona fide adipose-resident MDSCs. Systemic MDSC depletion altered adipose immune cell dynamics in otherwise healthy mice, increasing the number of CD4+ effector memory T cells and modifying the surface markers expressed by adipose-resident macrophages. In addition, transcription of various immunomodulatory cytokines was clearly dysregulated in the adipose of MDSC-depleted animals compared with controls. Altogether, our findings indicate that there is a population of bona fide MDSCs in the adipose of otherwise healthy mice that actively contribute to the health and immune homeostasis of this tissue.


Assuntos
Tecido Adiposo/imunologia , Homeostase/imunologia , Células Supressoras Mieloides , Animais , Antígeno CD11b , Citocinas , Ativação Linfocitária , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/imunologia , Linfócitos T
3.
J Leukoc Biol ; 106(6): 1349-1358, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31568613

RESUMO

Dendritic cells (DC) from diabetes-prone NOD mice and patients with type 1 diabetes (T1D) produce excess IL-12 that drives development of ß-cell-destroying IFN-γ-producing T cells. The molecular mechanisms that control IL-12 production in T1D are unclear. In this study, we report that ß-catenin, a multifunctional protein involved in inflammation, is dramatically increased in DC from NOD mice. We further investigated the mechanisms leading to accumulation of ß-catenin in NOD DC and its role in the inflammatory pathogenic responses associated with T1D. Hyperphosphorylation of ß-catenin at a stabilizing residue, serine 552, mediated by activation of Akt, appears to lead to ß-catenin accumulation in NOD DC. Elevated ß-catenin in DC correlated with IL-12 production and induction of IFN-γ-producing CD4 cells. On the one hand, knockdown/inhibition of ß-catenin significantly reduced NOD DC production of IL-12 and their ability to induce IFN-γ-producing CD4 cells. On the other hand, overexpression of ß-catenin in control DC resulted in increased IL-12 production and induction of IFN-γ-production in T cells. Additionally, we found that ß-catenin inhibitors decreased NF-κB activation in NOD DC and IFN-γ production by NOD T cells in vivo. These data strongly suggest that accumulation of ß-catenin in DC from NOD mice drives IL-12 production, and consequently, development of pathogenic IFN-γ-producing T cells. Targeting the defect responsible for ß-catenin accumulation and subsequent overproduction of pro-inflammatory cytokines by NOD DC could be an effective therapeutic strategy for the prevention and/or treatment of T1D.


Assuntos
Células Dendríticas/metabolismo , Interferon gama/biossíntese , Interleucina-12/biossíntese , Subpopulações de Linfócitos T/metabolismo , beta Catenina/metabolismo , Animais , Biomarcadores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos NOD , NF-kappa B/metabolismo , Fosforilação , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , beta Catenina/antagonistas & inibidores
4.
Biochem Biophys Res Commun ; 437(1): 29-34, 2013 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-23792098

RESUMO

Shwachman Diamond syndrome (SDS) is an inherited bone marrow failure syndrome typically characterized by neutropenia, exocrine pancreas dysfunction, metaphyseal chondrodysplasia, and predisposition to myelodysplastic syndrome and leukemia. SBDS, the gene affected in most cases of SDS, encodes a protein known to influence many cellular processes including ribosome biogenesis, mitotic spindle assembly, chemotaxis, and the regulation of reactive oxygen species production. The best characterized role for the SBDS protein is in the production of functional 60S ribosomal subunits. Given that a reduction in functional 60S subunits could impact on the translational output of cells depleted of SBDS we analyzed protein synthesis in yeast cells lacking SDO1, the ortholog of SBDS. Cells lacking SDO1 selectively increased the synthesis of POR1, the ortholog of mammalian VDAC1 a major anion channel of the mitochondrial outer membrane. Further studies revealed the cells lacking SDO1 were compromised in growth on non-fermentable carbon sources suggesting mitochondrial function was impaired. These observations prompted us to examine mitochondrial function in human cells where SBDS expression was reduced. Our studies indicate that reduced expression of SBDS decreases mitochondrial membrane potential and oxygen consumption and increases the production of reactive oxygen species. These studies indicate that mitochondrial function is also perturbed in cells expressing reduced amounts of SBDS and indicate that disruption of mitochondrial function may also contribute to SDS pathophysiology.


Assuntos
Doenças da Medula Óssea/metabolismo , Doenças da Medula Óssea/patologia , Insuficiência Pancreática Exócrina/metabolismo , Insuficiência Pancreática Exócrina/patologia , Lipomatose/metabolismo , Lipomatose/patologia , Mitocôndrias/metabolismo , Modelos Biológicos , Saccharomyces cerevisiae/metabolismo , Carbono/farmacologia , Linhagem Celular , Fermentação/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Proteínas/metabolismo , Proteômica , Espécies Reativas de Oxigênio/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/metabolismo , Síndrome de Shwachman-Diamond
5.
Pediatr Blood Cancer ; 60(2): 281-6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22997148

RESUMO

BACKGROUND: Shwachman-Diamond syndrome (SDS), associated with SBDS mutations, is characterized by pancreatic exocrine dysfunction and marrow failure. Sdo1, the yeast ortholog of SBDS, is implicated in maturation of the 60S ribosomal subunit, with delayed export of 60S-like particles from the nucleoplasm when depleted. Sdo1 is needed for release of the anti-subunit association factor Tif6 from 60S subunits, and Tif6 may not be recycled to the nucleus when Sdo1 is absent. METHODS: To clarify the role of SBDS in human ribosome function, TF-1 erythroleukemia and A549 lung carcinoma cells were transfected with vectors expressing RNAi against SBDS. RESULTS: Growth and hematopoietic colony forming potential of TF-1 knockdown cells were markedly hindered when compared to controls. To analyze the effect of SBDS on 60S subunit maturation in A549 cells, subunit localization was assessed by transfection with a vector expressing a fusion between human RPL29 and GFP: we found a higher percentage of SBDS-depleted cells with nuclear localization of 60S subunits. Polysome analysis of TF-1 knockdown cells showed a decrease in free 60S and 80S subunits. We also analyzed the levels of eIF6 (human ortholog of Tif6) following near-complete knockdown of SBDS in TF-1 cells and found an approximately 20% increase in the amount of eIF6 associated with the 60S subunit. CONCLUSIONS: We conclude that knockdown of SBDS leads to growth inhibition and defects in ribosome maturation, suggesting a role for wild-type SBDS in nuclear export of pre-60S subunits. Furthermore, knockdown of SBDS may interfere with eIF6 recycling.


Assuntos
Hematopoese/fisiologia , Proteínas/metabolismo , Ribossomos/fisiologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Silenciamento de Genes , Humanos , Proteínas/genética , RNA Interferente Pequeno , Transfecção
6.
Front Microbiol ; 2: 180, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21922015

RESUMO

Our immune system has evolved to recognize and eradicate pathogenic microbes. However, we have a symbiotic relationship with multiple species of bacteria that occupy the gut and comprise the natural commensal flora or microbiota. The microbiota is critically important for the breakdown of nutrients, and also assists in preventing colonization by potentially pathogenic bacteria. In addition, the gut commensal bacteria appear to be critical for the development of an optimally functioning immune system. Various studies have shown that individual species of the microbiota can induce very different types of immune cells (e.g., Th17 cells, Foxp3(+) regulatory T cells) and responses, suggesting that the composition of the microbiota can have an important influence on the immune response. Although the microbiota resides in the gut, it appears to have a significant impact on the systemic immune response. Indeed, specific gut commensal bacteria have been shown to affect disease development in organs other than the gut, and depending on the species, have been found to have a wide range of effects on diseases from induction and exacerbation to inhibition and protection. In this review, we will focus on the role that the gut microbiota plays in the development and progression of inflammatory/autoimmune disease, and we will also touch upon its role in allergy and cancer.

7.
J Immunol ; 186(9): 5217-26, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21422248

RESUMO

The PI3K pathway and its regulation of mammalian target of rapamycin complex 1 (mTORC1) and glycogen synthase kinase 3 (GSK3) play pivotal roles in controlling inflammation. In this article, we show that mTORC1 and GSK3-ß converge and that the capacity of mTORC1 to affect the inflammatory response is due to the inactivation of GSK3-ß. Inhibition of mTORC1 attenuated GSK3 phosphorylation and increased its kinase activity. Immunoprecipitation and in vitro kinase assays demonstrated that GSK3-ß associated with a downstream target of mTORC1, p85S6K, and phosphorylated GSK3-ß. Inhibition of S6K1 abrogated the phosphorylation of GSK3-ß while increasing and decreasing the levels of IL-12 and IL-10, respectively, in LPS-stimulated monocytes. In contrast, the direct inhibition of GSK3 attenuated the capacity of S6K1 inhibition to influence the levels of IL-10 and IL-12 produced by LPS-stimulated cells. At the transcriptional level, mTORC1 inhibition reduced the DNA binding of CREB and this effect was reversed by GSK3 inhibition. As a result, mTORC1 inhibition increased the levels of NF-κB p65 associated with CREB-binding protein. Inhibition of NF-κB p65 attenuated rapamycin's ability to influence the levels of pro- or anti-inflammatory cytokine production in monocytes stimulated with LPS. These studies identify the molecular mechanism by which mTORC1 affects GSK3 and show that mTORC1 inhibition regulates pro- and anti-inflammatory cytokine production via its capacity to inactivate GSK3.


Assuntos
Quinase 3 da Glicogênio Sintase/imunologia , Imunidade Inata/imunologia , Proteínas/imunologia , Transdução de Sinais/imunologia , Western Blotting , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Separação Celular , Citocinas/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Expressão Gênica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Imunoprecipitação , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Monócitos/imunologia , Monócitos/metabolismo , Complexos Multiproteicos , Proteínas/metabolismo , Serina-Treonina Quinases TOR
8.
J Immunol ; 181(10): 6797-802, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18981097

RESUMO

TLR 4 stimulation of innate immune cells induces a MyD88-independent signaling pathway that leads to the production of IFN-beta. In this study, we demonstrate glycogen synthase kinase 3-beta (GSK3-beta) plays a fundamental role in this process. Suppression of GSK3-beta activity by either pharmacological inhibition, small interfering RNA-mediated gene silencing, or ectopic expression of a kinase-dead GSK3-beta mutant enhanced IFN-beta production by TLR4-stimulated macrophages. Conversely, ectopic expression of a constitutively active GSK3-beta mutant severely attenuated IFN-beta production. GSK3-beta was found to negatively control the cellular levels of the transcription factor c-Jun and its nuclear association with ATF-2. Small interfering RNA-mediated knockdown of c-Jun levels abrogated the ability of GSK3-beta inhibition to augment IFN-beta, demonstrating that the ability of GSK3 to control IFN-beta production was due to its ability to regulate c-Jun levels. The ability of GSK3 inhibition to control IFN-beta production was confirmed in vivo as mice treated with a GSK3 inhibitor exhibited enhanced systemic levels of IFN-beta upon LPS challenge. These findings identify a novel regulatory pathway controlling IFN-beta production by TLR4-stimulated innate immune cells.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Interferon beta/biossíntese , Macrófagos/imunologia , Receptor 4 Toll-Like/imunologia , Fator 2 Ativador da Transcrição/imunologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Western Blotting , Quinase 3 da Glicogênio Sintase/imunologia , Glicogênio Sintase Quinase 3 beta , Interferon beta/imunologia , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Fosforilação , Proteínas Proto-Oncogênicas c-jun/imunologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Mol Immunol ; 44(11): 2884-92, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17324464

RESUMO

Activation induced cell death (AICD) via Fas/FasL is the primary homeostatic molecular mechanism employed by the immune system to control activated T-cell responses and promote tolerance to self-antigens. We herein investigated the ability of a novel multimeric form of FasL chimeric with streptavidin (SA-FasL) having potent apoptotic activity to induce apoptosis in diabetogenic T cells and modulate insulin-dependent type 1 diabetes (IDDM) in an adoptive transfer model. Diabetogenic splenocytes from NOD/Lt females were co-cultured in vitro with SA-FasL, SA control protein, or alone without protein, and adoptively transferred into NOD/Lt-Rag1(null) recipients for diabetes development. All animals receiving control (Alone: n=16 or SA: n=17) cells developed diabetes on average by 6 weeks, whereas animals receiving SA-FasL-treated (n=25) cells exhibited significantly delayed progression (p<.001) and decreased incidence (70%). This effect was associated with an increase in CD4(+)CD25(+) T cells and correlated with FoxP3 expression in pancreatic lymph nodes. Extracorporeal treatment of peripheral blood lymphocytes using SA-FasL during disease onset represents a novel approach that may alter the ability of pathogenic T cells to mediate diabetes and have therapeutic utility in clinical management of IDDM.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Proteína Ligante Fas/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Apoptose/imunologia , Antígenos CD4/imunologia , Células Cultivadas , Técnicas de Cocultura , Diabetes Mellitus Tipo 1/etiologia , Feminino , Fatores de Transcrição Forkhead/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos NOD , Ratos , Proteínas Recombinantes de Fusão/imunologia , Estreptavidina/imunologia
10.
J Immunol ; 177(10): 6962-73, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17082611

RESUMO

A single intratumoral injection of IL-12 and GM-CSF-loaded slow-release microspheres induces T cell-dependent eradication of established primary and metastatic tumors in a murine lung tumor model. To determine how the delivery of cytokines directly to the microenvironment of a tumor nodule induces local and systemic antitumor T cell activity, we characterized therapy-induced phenotypic and functional changes in tumor-infiltrating T cell populations. Analysis of pretherapy tumors demonstrated that advanced primary tumors were infiltrated by CD4+ and CD8+ T cells with an effector/memory phenotype and CD4+CD25+Foxp3+ T suppressor cells. Tumor-associated effector memory CD8+ T cells displayed impaired cytotoxic function, whereas CD4+CD25+Foxp3+ cells effectively inhibited T cell proliferation demonstrating functional integrity. IL-12/GM-CSF treatment promoted a rapid up-regulation of CD43 and CD69 on CD8+ effector/memory T cells, augmented their ability to produce IFN-gamma, and restored granzyme B expression. Importantly, treatment also induced a concomitant and progressive loss of T suppressors from the tumor. Further analysis established that activation of pre-existing effector memory T cells was short-lived and that both the effector/memory and the suppressor T cells became apoptotic within 4 days of treatment. Apoptotic death of pre-existing effector/memory and suppressor T cells was followed by infiltration of the tumor with activated, nonapoptotic CD8+ effector T lymphocytes on day 7 posttherapy. Both CD8+ T cell activation and T suppressor cell purge were mediated primarily by IL-12 and required IFN-gamma. This study provides important insight into how local IL-12 therapy alters the immunosuppressive tumor milieu to one that is immunologically active, ultimately resulting in tumor regression.


Assuntos
Adenocarcinoma Bronquioloalveolar/terapia , Apoptose/imunologia , Linfócitos T CD8-Positivos/patologia , Movimento Celular/imunologia , Memória Imunológica , Interleucina-12/administração & dosagem , Neoplasias Pulmonares/terapia , Linfócitos do Interstício Tumoral/patologia , Linfócitos T Reguladores/imunologia , Adenocarcinoma Bronquioloalveolar/imunologia , Adenocarcinoma Bronquioloalveolar/patologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/uso terapêutico , Morte Celular/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Injeções Intralesionais , Interleucina-12/uso terapêutico , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Microesferas , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia
11.
Diabetes ; 55(7): 2098-105, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16804081

RESUMO

Various defects in antigen-presenting cells (APCs) and T-cells, including regulatory cells, have been associated with type 1 diabetes development in NOD mice. CD4(+)CD25(+) regulatory cells play a crucial role in controlling various autoimmune diseases, and a deficiency in their number or function could be involved in disease development. The current study shows that NOD mice had fewer CD4(+)CD25(+) regulatory cells, which expressed normal levels of glucocorticoid-induced tumor necrosis factor receptor and cytotoxic T-lymphocyte-associated antigen-4. We have also found that NOD CD4(+)CD25(+) cells regulate poorly in vitro after stimulation with anti-CD3 and NOD APCs in comparison with B6 CD4(+)CD25(+) cells stimulated with B6 APCs. Surprisingly, stimulation of NOD CD4(+)CD25(+) cells with B6 APCs restored regulation, whereas with the reciprocal combination, NOD APCs failed to activate B6 CD4(+)CD25(+) cells properly. Interestingly, APCs from disease-free (>30 weeks of age), but not diabetic, NOD mice were able to activate CD4(+)CD25(+) regulatory function in vitro and apparently in vivo because only spleens of disease-free NOD mice contained potent CD4(+)CD25(+) regulatory cells that prevented disease development when transferred into young NOD recipients. These data suggest that the failure of NOD APCs to activate CD4(+)CD25(+) regulatory cells may play an important role in controlling type 1 diabetes development in NOD mice.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Receptores de Interleucina-2/imunologia , Linfócitos T/imunologia , Animais , Diabetes Mellitus Tipo 1/genética , Feminino , Citometria de Fluxo , Imunofenotipagem , Ativação Linfocitária , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Reação em Cadeia da Polimerase , Estado Pré-Diabético/genética , Estado Pré-Diabético/imunologia , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação
12.
Immunol Res ; 30(2): 155-70, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15477657

RESUMO

Tolerogenic antigen-presenting cells (APCs) are attractive agents for the treatment of autoimmune and inflammatory diseases that are mediated, at least in part, by antigen-specific autoreactive T cells. Transforming growth factor-beta (TGF-beta)-treated antigen-presenting cells induce a very potent form of tolerance in mice. One unique feature of this simple and elegant method of tolerance induction is that it is equally potent in both primed and naive mice, an important consideration for the development of a therapy that will be effective against an established disease. In this model, F4/80+ peritoneal exudate cells (macrophages) cultured with antigen and TGF-beta2 injected iv induce populations of regulatory T cells that mediate long-lasting antigen-specific tolerance in mice. The mechanisms that are involved in the induction of tolerance by TGF-beta-treated APCs are very complex and require the interaction of a variety of cell types, as well as soluble and membrane-bound factors. In this review, we summarize the existing data and present new data concerning the induction and effector mechanisms associated with TGF-beta-treated APC-induced tolerance. An understanding of these mechanisms will provide very important information for the design of effective strategies for the treatment of a variety of diseases that are mediated by pathogenic T cells.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Tolerância Imunológica , Macrófagos Peritoneais/imunologia , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Células Apresentadoras de Antígenos/transplante , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Doenças Autoimunes/terapia , Humanos , Imunoterapia Adotiva , Inflamação/imunologia , Inflamação/patologia , Inflamação/terapia , Macrófagos Peritoneais/transplante , Camundongos , Linfócitos T/patologia
13.
Int Immunol ; 16(5): 697-706, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15096489

RESUMO

Transforming growth factor (TGF)-beta-treated antigen-presenting cells [(APC) adherent peritoneal exudate cells] induce a profound tolerance in primed mice that is thought to be mediated by regulatory T cells induced in the spleen. In the current study, we investigated the mechanism(s) involved in tolerance induced in primed mice by TGF-beta-treated APC. Interestingly, TGF-beta-treated APC from class II knockout mice were unable to mediate tolerance in primed mice and failed to induce not only CD4, but also CD8 regulatory T cells. However, the results of several experiments indicated that it was the CD8 regulatory T cells that were required for tolerance induced in primed mice. Using neutralizing antibody, we found that TGF-beta-treated APC-induced CD8 regulatory T cells did not suppress effector T cell function in vivo through the production of IL-4, TGF-beta or IL-10. On the other hand, our data showed that the Fas-Fas ligand (FasL) pathway was involved in this form of tolerance since TGF-beta-treated APC could not mediate tolerance in primed FasL-deficient mice and CD8 T cells from FasL-deficient mice were unable to suppress effector T cell responses. Moreover, the targets of FasL-mediated suppression were found to be the effector T cells as suggested by the data showing that Fas-deficient effector T cells were not susceptible to suppression mediated by CD8 regulatory T cells induced by TGF-beta-treated APC. In conclusion, our data indicate that TGF-beta-treated APC effect tolerance in primed mice via a Fas-FasL-mediated mechanism that requires CD8 cells.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Tolerância Imunológica/imunologia , Glicoproteínas de Membrana/fisiologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Proteína Ligante Fas , Feminino , Genes MHC da Classe II/genética , Hipersensibilidade Tardia/imunologia , Tolerância Imunológica/genética , Imunização , Interleucina-10/imunologia , Interleucina-4/imunologia , Depleção Linfocítica , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Ovalbumina/imunologia
14.
Int Immunol ; 15(8): 945-53, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12882832

RESUMO

Intravenous injection of transforming growth factor (TGF-)-beta-treated antigen-presenting cells (APC) pulsed with antigen induces antigen-specific tolerance in both naive and previously primed mice. Although TGF-beta-treated APC-induced tolerance is associated with induction of regulatory T cells and impaired delayed-type hypersensitivity (DTH) responses, the specific mechanisms that mediate this tolerance are not currently known. The goal of the present report was to study the mechanisms involved in TGF-beta-treated APC-induced tolerance by determining the fate of the antigen-specific effector T cells that are regulated. Using a well-characterized system that allows tracking of small numbers of TCR transgenic T cells, we have found that antigen-specific T cell expansion, either in vivo or in vitro, is inhibited in mice that have been injected with TGF-beta-treated APC. The failure of antigen-specific effector T cells to expand did not appear to be due to the induction of anergy, since carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled cells divided normally in response to antigen and adjuvant in vivo, and addition of exogenous IL-2 was unable to restore T cell expansion in in vitro assays. Interestingly, the percentage of CFSE-labeled cells was decreased after >7-8 divisions following culture in vitro, which correlated with a significant increase in cell death. Cell death was prevented and the ability to expand in vitro was restored by treatment with anti-Fas ligand (FasL) antibody. In conclusion, tolerance induced by TGF-beta-treated APC appears to be associated with deletion of antigen-specific T cells involving the Fas-FasL pathway.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Anergia Clonal/imunologia , Deleção Clonal/imunologia , Tolerância Imunológica/imunologia , Fator de Crescimento Transformador beta/imunologia , Transferência Adotiva , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Antígenos de Diferenciação de Linfócitos T/análise , Apoptose/imunologia , Linfócitos T CD4-Positivos/química , Morte Celular/imunologia , Divisão Celular/imunologia , Células Cultivadas , Proteína Ligante Fas , Feminino , Citometria de Fluxo/métodos , Fluoresceínas/análise , Interleucina-2/imunologia , Interleucina-2/farmacologia , Linfonodos/citologia , Ativação Linfocitária/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Ovalbumina/imunologia , Ovalbumina/farmacologia , Fragmentos de Peptídeos/genética , Cavidade Peritoneal/citologia , Receptores de Antígenos de Linfócitos T/análise , Receptores de Antígenos de Linfócitos T/genética , Succinimidas/análise , Tioglicolatos/farmacologia , Fator de Crescimento Transformador beta/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA