RESUMO
The B cell response to Ehrlichia muris is dominated by plasmablasts (PBs), with few-if any-germinal centers (GCs), yet it generates protective immunoglobulin M (IgM) memory B cells (MBCs) that express the transcription factor T-bet and harbor V-region mutations. Because Ehrlichia prominently infects the liver, we investigated the nature of liver B cell response and that of the spleen. B cells within infected livers proliferated and underwent somatic hypermutation (SHM). Vh-region sequencing revealed trafficking of clones between the spleen and liver and often subsequent local clonal expansion and intraparenchymal localization of T-bet+ MBCs. T-bet+ MBCs expressed MBC subset markers CD80 and PD-L2. Many T-bet+ MBCs lacked CD11b or CD11c expression but had marginal zone (MZ) B cell phenotypes and colonized the splenic MZ, revealing T-bet+ MBC plasticity. Hence, liver and spleen are generative sites of B cell responses, and they include V-region mutation and result in liver MBC localization.
Assuntos
Linfócitos B/imunologia , Ehrlichia/imunologia , Ehrlichiose/imunologia , Imunoglobulina M/imunologia , Fígado/imunologia , Baço/imunologia , Animais , Antígeno B7-1/biossíntese , Região Variável de Imunoglobulina/genética , Memória Imunológica/imunologia , Fígado/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína 2 Ligante de Morte Celular Programada 1/biossíntese , Hipermutação Somática de Imunoglobulina/genética , Baço/citologia , Proteínas com Domínio T/metabolismoRESUMO
Although antigen recognition mediated by the T cell receptor (TCR) influences many facets of Foxp3(+) regulatory T (Treg) cell biology, including development and function, the cell types that present antigen to Treg cells in vivo remain largely undefined. By tracking a clonal population of Aire-dependent, prostate-specific Treg cells in mice, we demonstrated an essential role for dendritic cells (DCs) in regulating organ-specific Treg cell biology. We have shown that the thymic development of prostate-specific Treg cells required antigen presentation by DCs. Moreover, Batf3-dependent CD8α(+) DCs were dispensable for the development of this clonotype and had negligible impact on the polyclonal Treg cell repertoire. In the periphery, CCR7-dependent migratory DCs coordinated the activation of organ-specific Treg cells in the prostate-draining lymph nodes. Our results demonstrate that the development and peripheral regulation of organ-specific Treg cells are dependent on antigen presentation by DCs, implicating DCs as key mediators of organ-specific immune tolerance.
Assuntos
Apresentação de Antígeno/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/genética , Células Dendríticas/imunologia , Próstata/imunologia , Proteínas Repressoras/genética , Tolerância a Antígenos Próprios/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Antígeno B7-2/biossíntese , Antígeno B7-2/genética , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Antígenos CD8/metabolismo , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Próstata/citologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores CCR7/metabolismo , Proteínas Repressoras/imunologia , Linfócitos T Reguladores/citologia , Fatores de Transcrição/metabolismo , Proteína AIRERESUMO
BATF3-deficient mice that lack CD8+ dendritic cells (DCs) showed an exacerbation of chronic graft-versus-host disease (cGVHD), including T follicular helper (Tfh) cell and autoantibody responses, whereas mice carrying the Sle2c2 lupus-suppressive locus with a mutation in the G-CSFR showed an expansion of CD8+ DCs and a poor mobilization of plasmacytoid DCs (pDCs) and responded poorly to cGVHD induction. Here, we investigated the contribution of CD8+ DCs and pDCs to the humoral response to protein immunization, where CD8neg DCs are thought to represent the major inducers. Both BATF3-/- and Sle2c2 mice had reduced humoral and germinal center (GC) responses compared with C57BL/6 (B6) controls. We showed that B6-derived CD4+ DCs are the major early producers of IL-6, followed by CD4-CD8- DCs. Surprisingly, IL-6 production and CD80 expression also increased in CD8+ DCs after immunization, and B6-derived CD8+ DCs rescued Ag-specific adaptive responses in BATF3-/- mice. In addition, inflammatory pDCs (ipDCs) produced more IL-6 than all conventional DCs combined. Interestingly, G-CSFR is highly expressed on pDCs. G-CSF expanded pDC and CD8+ DC numbers and IL-6 production by ipDCs and CD4+ DCs, and it improved the quality of Ab response, increasing the localization of Ag-specific T cells to the GC. Finally, G-CSF activated STAT3 in early G-CSFR+ common lymphoid progenitors of cDCs/pDCs but not in mature cells. In conclusion, we showed a multilayered role of DC subsets in priming Tfh cells in protein immunization, and we unveiled the importance of G-CSFR signaling in the development and function pDCs.
Assuntos
Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/imunologia , Células Progenitoras Linfoides/citologia , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Células T Auxiliares Foliculares/imunologia , Transferência Adotiva , Animais , Autoanticorpos/imunologia , Antígeno B7-1/biossíntese , Fatores de Transcrição de Zíper de Leucina Básica/genética , Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Diferenciação Celular/imunologia , Células Dendríticas/transplante , Feminino , Fator Estimulador de Colônias de Granulócitos/metabolismo , Interleucina-6/biossíntese , Ativação Linfocitária/imunologia , Células Progenitoras Linfoides/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Proteínas Repressoras/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/imunologiaRESUMO
Atg16L1 mediates the cellular degradative process of autophagy and is considered a critical regulator of inflammation based on its genetic association with inflammatory bowel disease. Here we find that Atg16L1 deficiency leads to an exacerbated graft-versus-host disease (GVHD) in a mouse model of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Atg16L1-deficient allo-HSCT recipients with GVHD displayed increased T cell proliferation due to increased dendritic cell (DC) numbers and costimulatory molecule expression. Reduced autophagy within DCs was associated with lysosomal abnormalities and decreased amounts of A20, a negative regulator of DC activation. These results broaden the function of Atg16L1 and the autophagy pathway to include a role in limiting a DC-mediated response during inflammatory disease, such as GVHD.
Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas de Transporte/imunologia , Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/imunologia , Animais , Autofagia/imunologia , Proteínas Relacionadas à Autofagia , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Antígenos CD40/biossíntese , Proteínas de Transporte/genética , Proliferação de Células , Células Cultivadas , Colite/imunologia , Cisteína Endopeptidases/biossíntese , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Transplante de Células-Tronco Hematopoéticas , Proteínas de Homeodomínio/genética , Proteínas Imediatamente Precoces/biossíntese , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Ativação Linfocitária/imunologia , Lisossomos/patologia , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Transplante Homólogo , Proteína 3 Induzida por Fator de Necrose Tumoral alfaRESUMO
The prognosis of autoimmune thyroid diseases (AITDs), such as Graves' disease (GD) and Hashimoto's disease (HD), are difficult to predict. Both CD80 and CD86 costimulatory signals promote T cell activation in cooperation with T cell receptor signal. To clarify whether any association between CD80 and CD86 and the pathogenesis of AITD exist, we examined the expressions and gene polymorphisms of CD80 and CD86. We examined the expressions of CD80 and CD86 proteins on peripheral blood cells by flowcytometry and genotyped CD80 and CD86 gene polymorphisms by PCR-RFLP and Taqman PCR methods. In the analysis of the Blymphocytes elevated CD80+ cells (>8%) were found more often in the patients than in control subjects, and also it was more frequent in patients with intractable GD than in those with GD in remission (p= .0176). The mean fluorescence intensity of CD86 expression on monocytes was higher in GD and HD patients than in control subjects (p= <0.0001 and p= .0017, respectively). CD80 rs1599795 T allele carriers were more frequent in patients with severe HD than in those with mild HD. CD86 rs2715267 AA genotype was more frequent in HD patients than in controls. In conclusion, the expressions of CD80 on Bcells and of CD86 on monocytes were increased in peripheral blood from patients with AITD, especially in severe cases, and their gene polymorphisms are associated with the susceptibility and the severity of HD.
Assuntos
Antígeno B7-1/genética , Antígeno B7-2/genética , Doença de Graves/genética , Doença de Hashimoto/genética , Adulto , Linfócitos B/metabolismo , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Feminino , Predisposição Genética para Doença/genética , Doença de Graves/metabolismo , Doença de Hashimoto/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Polimorfismo de Nucleotídeo ÚnicoRESUMO
Manipulation of costimulatory and surface molecules that shape the extent of immune responses by Leishmania is suggested as one of the mechanisms of evading the host's defences. The experiments reported here were designed to evaluate the expressions of CD11b, CD11c, CD14, CD18, CD54, CD80, CD86, CD206, MHC class II and TLR-2 (Toll-like receptor 2) in human macrophages infected with L. amazonensis. Phenotypic evaluation revealed a negative modulation in CD11b, CD11c, CD14, CD18, CD54 and MHC class II molecules, depending on the level of infection. The results showed that as early as 1 hour after infection no reduction in marker expression occurs, whereas after 24 hours, downregulation of these molecules was observed in macrophages. No significant changes were observed in the expressions of CD80, CD86, CD206 and TLR2. Evidence of the differential modulation of markers expression and that after parasite uptake no reduction in surface marker expression occurs indicates that parasite internalization is not involved in the phenomena of down-modulation.
Assuntos
Antígenos CD/biossíntese , Leishmania mexicana/imunologia , Leishmaniose Cutânea/imunologia , Macrófagos/imunologia , Macrófagos/parasitologia , Adulto , Animais , Antígenos CD/imunologia , Antígenos de Superfície/biossíntese , Antígenos de Superfície/imunologia , Antígeno B7-1/biossíntese , Antígeno CD11b/biossíntese , Células Cultivadas , Antígenos de Histocompatibilidade Classe II/biossíntese , Humanos , Leishmaniose Cutânea/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor 2 Toll-Like/biossínteseRESUMO
MicroRNAs (miRNAs) are short noncoding RNAs that regulate a broad spectrum of biological processes, including immune responses. Although the contributions of miRNAs to the function of immune cells are beginning to emerge, their specific roles remain largely unknown. IL-33 plays an important role in macrophage activation for innate host defense and proinflammatory responses. In this study, we report that miR-487b can suppress the levels of mRNA and protein for IL-33 during the differentiation of bone marrow-derived macrophages (BMDMs). This results in inhibition of IL-33-induced expression of Ag-presenting and costimulatory molecules and proinflammatory mediators. A luciferase assay showed that miR-487b binds to the IL-33 3'-untranslated region. We also confirmed that IL-33 directly promotes the activation of BMDMs by increasing the expression of MHC class I, MHC class II, CD80/CD86, and inducible NO synthase (iNOS) in a dose-dependent manner. Exposure of BMDMs to the TLR4 ligand, LPS, decreased miR-487b expression, increased IL-33 transcript levels, and induced the production of proinflammatory mediators (e.g., iNOS, IL-1ß, IL-6, and TNF-α). Treatment with a specific inhibitor of miR-487b function also resulted in increased levels of IL-33 mRNA, which augmented LPS-induced expression of these inflammatory mediators in macrophages. Collectively, our results indicate that miR-487b plays a negative regulatory role in macrophages by controlling the levels of IL-33 transcript and protein to fine-tune innate immune host defense and proinflammatory responses of these cells. Thus, miR-487b plays an important role in the regulation of macrophage homeostasis and activation by targeting IL-33 transcripts.
Assuntos
Interleucina-33/biossíntese , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , MicroRNAs/genética , Regiões 3' não Traduzidas/genética , Animais , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Sítios de Ligação/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe II/biossíntese , Interleucina-1beta/biossíntese , Interleucina-33/genética , Interleucina-33/imunologia , Interleucina-6/biossíntese , Lipopolissacarídeos , Ativação de Macrófagos/genética , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/biossínteseRESUMO
Notch signaling is involved in regulating TLR-mediated responses in activated macrophages. In this study, we investigated the impact of Notch signaling in macrophages in an experimental autoimmune encephalomyelitis (EAE) model. To examine the impact of deficiency in Notch signaling in activated macrophages in EAE, an adoptive transfer of activated macrophages derived from Notch1(fl/fl) × Mx1cre(+/-) (Notch1 knockout [N1KO]) or CSL/Rbp-jκ(fl/fl) × Mx1cre(+/-) (CSL/RBP-Jκ KO) mice was performed prior to induction of EAE. Mice receiving activated N1KO macrophages showed decreased severity of EAE compared with mice receiving wild-type or CSL/RBP-Jκ KO macrophages. In vitro restimulation of splenocytes by myelin oligodendrocyte glycoprotein 35-55 peptide from these mice revealed that cells from mice receiving N1KO macrophages produced significantly less IL-17 compared with the control mice, whereas IFN-γ production was similar in both groups. We found that activated N1KO, but not CSL/RBP-Jκ KO, macrophages produced less IL-6 and had lower CD80 expression compared with wild-type and did not exhibit any defect in IL-12p40/70 production, whereas activated macrophages from CSL/RBP-Jκ KO mice phenocopied γ-secretase inhibitor treatment for reduced IL-12p40/70 production. Furthermore, the nuclear translocation of the NF-κB subunit c-Rel was compromised in γ-secretase inhibitor-treated and CSL/RBP-Jκ KO but not N1KO macrophages. These results suggest that Notch1 and CSL/RBP-Jκ in macrophages may affect the severity of EAE differently, possibly through modulating IL-6 and CD80 expression, which is involved in the Th17 but not Th1 response.
Assuntos
Encefalomielite Autoimune Experimental/imunologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Macrófagos/imunologia , Receptor Notch1/genética , Células Th17/imunologia , Transferência Adotiva , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Antígeno B7-1/biossíntese , Células Cultivadas , Técnicas de Cocultura , Feminino , Deleção de Genes , Interferon gama/biossíntese , Subunidade p40 da Interleucina-12/biossíntese , Interleucina-17/biossíntese , Interleucina-6/biossíntese , Macrófagos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-rel/metabolismo , Transdução de Sinais/imunologiaRESUMO
Cellular FLIP (c-FLIP) specifically inhibits caspase-8 and suppresses death receptor-induced apoptosis. c-FLIP has also been reported to transmit activation signals. In this study, we report a novel function of c-FLIP involving inhibition of myeloid cell activation through antagonizing the selective innate signaling pathway. We found that conditional knockout of c-FLIP in dendritic cells (DCs) led to neutrophilia and splenomegaly. Peripheral DC populations, including CD11b(+) conventional DCs (cDCs), CD8(+) cDCs, and plasmacytoid DCs, were not affected by c-FLIP deficiency. We also found that c-FLIP knockout cDCs, plasmacytoid DCs, and bone marrow-derived DCs (BMDCs) displayed enhanced production of TNF-α, IL-2, or G-CSF in response to stimulation of TLR4, TLR2, and dectin-1. Consistent with the ability of c-FLIP to inhibit the activation of p38 MAPK, the enhanced activation of c-FLIP-deficient BMDCs could be partly linked to an elevated activation of p38 MAPK after engagement of innate receptors. Increased activation was also found in c-FLIP(+/-) macrophages. Additionally, the increased activation in c-FLIP-deficient DCs was independent of caspase-8. Our results reveal a novel inhibitory role of c-FLIP in myeloid cell activation and demonstrate the unexpected anti-inflammatory activity of c-FLIP. Additionally, our observations suggest that cancer therapy targeting c-FLIP downregulation may facilitate DC activation and increase T cell immunity.
Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/imunologia , Caspase 8/imunologia , Células Dendríticas/imunologia , Células Mieloides/imunologia , Animais , Anti-Inflamatórios , Apresentação de Antígeno/imunologia , Apoptose/imunologia , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Células da Medula Óssea/imunologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Caspase 8/genética , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Fator Estimulador de Colônias de Granulócitos/biossíntese , Antígenos de Histocompatibilidade Classe II/biossíntese , Inflamação/imunologia , Interleucina-2/biossíntese , Lectinas Tipo C/imunologia , Contagem de Leucócitos , Camundongos , Camundongos Knockout , Células Mieloides/citologia , Neutrófilos/citologia , Neutrófilos/imunologia , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/imunologia , Esplenomegalia/imunologia , Linfócitos T/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismoRESUMO
The incidence of progressive kidney disease associated with diabetes continues to rise worldwide. Current standard therapy with angiotensin-converting enzyme inhibitors and/or angiotensin receptor blockers achieves only partial renoprotection, increasing the need for novel therapeutic approaches. Previous studies described B7-1 induction in podocytes of patients with proteinuria, including those with FSGS and type 2 diabetic nephropathy (DN). These findings sparked great excitement in the renal community, implying that abatacept, a costimulatory inhibitor that targets B7-1, could be a novel therapy for diabetic renal disease. Given previous concerns over the value of B7-1 immunostaining and the efficacy of abatacept in patients with recurrent FSGS after renal transplantation, we investigated B7-1 expression in human and experimental DN before embarking on clinical studies of the use of B7-1 targeting strategies to treat proteinuria in DN. Immunohistochemical analysis of kidney specimens using different antibodies revealed that B7-1 is not induced in podocytes of patients with DN, independent of disease stage, or BTBR ob/obmice, a model of type 2 diabetes. These results do not support the use of abatacept as a therapeutic strategy for targeting podocyte B7-1 for the prevention or treatment of DN.
Assuntos
Antígeno B7-1/biossíntese , Nefropatias Diabéticas/metabolismo , Podócitos/metabolismo , Animais , Antígeno B7-1/análise , Humanos , Masculino , Camundongos , Podócitos/químicaRESUMO
Capparis spinosa L. (C. spinosa) has been used as food and traditional medicine and shows anti-inflammatory and anti-oxidant activities. Here, we prepared the C. spinosa fruit ethanol extracts (CSEs) using different procedures and investigated the effects of CSE on the maturation of mouse bone marrow-derived dendritic cells (DCs) in the absence or presence of lipopolysaccharide (LPS). DC maturation and cytokine production were detected by flow cytometry and ELISA, respectively. We obtained three different CSEs and dissolved in water or DMSO, named CSE2W, CSEMW, CSE3W, CSE2D, CSEMD, and CSE3D, respectively. These CSEs showed different effects on DC maturation. CSEMW and CSEMD significantly increased the expressions of CD40, CD80, and CD86, in a dose-dependent manner. CSE2W and CSE2D also showed a modest effect on DC maturation, which enhanced the expression of CD40. CSE3W and CSE3D did not change DC maturation but suppressed LPS-induced DC maturation characterized by the decreased levels of CD40 and CD80. CSE3W and CSE3D also significantly inhibited the secretions of IL-12p40, IL-6, IL-1ß, and TNF-α induced by LPS. CSE3W further increased the level of IL-10 induced by LPS. Moreover, CSE3D suppressed LPS-induced DC maturation in vivo, which decreased the expressions of CD40 and CD80. These results suggested that CSE3W and CSE3D might be used to treat inflammatory diseases.
Assuntos
Capparis/química , Diferenciação Celular/efeitos dos fármacos , Células Dendríticas/citologia , Extratos Vegetais/farmacologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Células da Medula Óssea/citologia , Antígenos CD40/biossíntese , Células Dendríticas/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Etanol/metabolismo , Citometria de Fluxo , Frutas/metabolismo , Interleucina-10/metabolismo , Subunidade p40 da Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos , Medicina Tradicional do Leste Asiático , Camundongos , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/metabolismoRESUMO
Immunosuppressive CD11b(+) Gr-1(+) myeloid-derived suppressor cells (MDSCs) accumulate in the livers of tumor-bearing (TB) mice. We studied hepatic MDSCs in two murine models of immune-mediated hepatitis. Unexpectedly, treatment of TB mice with Concanavalin A (Con A) or α-galactosylceramide resulted in increased alanine aminotransferase (ALT) and aspartate aminotransferase (AST) serum levels in comparison to tumor-free mice. Adoptive transfer of hepatic MDSCs into naïve mice exacerbated Con A induced liver damage. Hepatic CD11b(+) Gr-1(+) cells revealed a polarized proinflammatory gene signature after Con A treatment. An IFN-γ-dependent upregulation of CD40 on hepatic CD11b(+) Gr-1(+) cells along with an upregulation of CD80, CD86, and CD1d after Con A treatment was observed. Con A treatment resulted in a loss of suppressor function by tumor-induced CD11b(+) Gr-1(+) MDSCs as well as enhanced reactive oxygen species (ROS)-mediated hepatotoxicity. CD40 knockdown in hepatic MDSCs led to increased arginase activity upon Con A treatment and lower ALT/AST serum levels. Finally, blockade of arginase activity in Cd40(-/-) tumor-induced myeloid cells resulted in exacerbation of hepatitis and increased ROS production in vivo. Our findings indicate that in a setting of acute hepatitis, tumor-induced hepatic MDSCs act as proinflammatory immune effector cells capable of killing hepatocytes in a CD40-dependent manner.
Assuntos
Antígenos CD40/metabolismo , Hepatite/imunologia , Células Mieloides/imunologia , Transferência Adotiva , Alanina Transaminase/sangue , Animais , Antígenos CD1d/biossíntese , Arginase/antagonistas & inibidores , Arginase/biossíntese , Arginase/metabolismo , Aspartato Aminotransferases/sangue , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Antígeno CD11b/metabolismo , Antígenos CD40/biossíntese , Antígenos CD40/genética , Linhagem Celular , Concanavalina A/farmacologia , Feminino , Galactosilceramidas/farmacologia , Hepatite/genética , Hepatócitos/imunologia , Hepatócitos/patologia , Fígado/citologia , Fígado/lesões , Neoplasias Hepáticas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitógenos/farmacologia , Células Mieloides/transplante , Espécies Reativas de Oxigênio/metabolismo , Receptores de Quimiocinas/metabolismoRESUMO
Lymph node (LN) stromal cells (LNSCs) form the functional structure of LNs and play an important role in lymphocyte survival and the maintenance of immune tolerance. Despite their broad spectrum of function, little is known about LNSC responses during microbial infection. In this study, we demonstrate that LNSC subsets display distinct kinetics following vaccinia virus infection. In particular, compared with the expansion of other LNSC subsets and the total LN cell population, the expansion of fibroblastic reticular cells (FRCs) was delayed and sustained by noncirculating progenitor cells. Notably, newly generated FRCs were preferentially located in perivascular areas. Viral clearance in reactive LNs preceded the onset of FRC expansion, raising the possibility that viral infection in LNs may have a negative impact on the differentiation of FRCs. We also found that MHC class II expression was upregulated in all LNSC subsets until day 10 postinfection. Genetic ablation of radioresistant stromal cell-mediated Ag presentation resulted in slower contraction of Ag-specific CD4(+) T cells. We propose that activated LNSCs acquire enhanced Ag-presentation capacity, serving as an extrinsic brake system for CD4(+) T cell responses. Disrupted function and homeostasis of LNSCs may contribute to immune deregulation in the context of chronic viral infection, autoimmunity, and graft-versus-host disease.
Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfonodos/citologia , Células Estromais/imunologia , Vaccinia virus/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/imunologia , Antígeno B7-1/biossíntese , Linfócitos T CD4-Positivos/transplante , Diferenciação Celular/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe II/biossíntese , Receptores de Hialuronatos/biossíntese , Tolerância Imunológica , Integrina beta1/biossíntese , Linfonodos/virologia , Receptor beta de Linfotoxina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parabiose , Receptores do Fator de Crescimento Derivado de Plaquetas/biossíntese , Transdução de Sinais/imunologia , Vacínia/imunologia , Molécula 1 de Adesão de Célula Vascular/biossínteseRESUMO
Deciphering the mechanisms that allow the induction of strong immune responses is crucial to developing efficient vaccines against infectious diseases and cancer. Based on the discovery that the adenylate cyclase from Bordetella pertussis binds to the CD11b/CD18 integrin, we developed a highly efficient detoxified adenylate cyclase-based vector (CyaA) capable of delivering a large variety of Ags to the APC. This vector allows the induction of protective and therapeutic immunity against viral and tumoral challenges as well as against transplanted tumors in the absence of any added adjuvant. Two therapeutic vaccine candidates against human papilloma viruses and melanoma have been developed recently, based on the CyaA vector, and are currently in clinical trials. We took advantage of one of these highly purified vaccines, produced under good manufacturing practice-like conditions, to decipher the mechanisms by which CyaA induces immune responses. In this study, we demonstrate that CyaA binds both human and mouse CD11b(+) dendritic cells (DCs) and induces their maturation, as shown by the upregulation of costimulatory and MHC molecules and the production of proinflammatory cytokines. Importantly, we show that DCs sense CyaA through the TLR4/Toll/IL-1R domain-containing adapter-inducing IFN-ß pathway, independent of the presence of LPS. These findings show that CyaA possesses the intrinsic ability to not only target DCs but also to activate them, leading to the induction of strong immune responses. Overall, this study demonstrates that Ag delivery to CD11b(+) DCs in association with TLR4/Toll/IL-1R domain-containing adapter-inducing IFN-ß activation is an efficient strategy to promote strong specific CD8(+) T cell responses.
Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Toxina Adenilato Ciclase/imunologia , Antígeno CD11b/imunologia , Células Dendríticas/imunologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-2/biossíntese , Bordetella pertussis/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/citologia , Feminino , Interferon beta/imunologia , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptores de Interleucina-1/imunologia , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/imunologia , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Tirosina/genéticaRESUMO
Chlamydia trachomatis and Chlamydia pneumoniae are important human pathogens that infect the urogenital/anorectal and respiratory tracts, respectively. Whilst the ability of these bacteria to infect epithelia is well defined, there is also considerable evidence of infection of leucocytes, including dendritic cells (DCs). Using a human dendritic cell line (MUTZ), we demonstrate that the infection and replication of chlamydiae inside DCs is species and serovar specific and that live infection with C. pneumoniae is required to upregulate costimulatory markers CD80, CD83 and human leucocyte antigen (HLA)-DR on MUTZ cells, as well as induce secretion of interleukin (IL)-2, IL-6, IL-8, IL-12 (p70), interferon-gamma and tumour necrosis factor-alpha Conversely, C. trachomatis serovar D failed to upregulate DC costimulatory markers, but did induce secretion of high concentrations of IL-8. Interestingly, we also observed that infection of MUTZ cells with C. pneumoniae or C. trachomatis serovar L2, whilst not replicative, remained infectious and upregulated lymph node migratory marker CCR7 mRNA. Taken together, these data confirm the findings of other groups using primary DCs and demonstrate the utility of MUTZ cells for further studies of chlamydial infection.
Assuntos
Infecções por Chlamydia/imunologia , Chlamydia trachomatis/imunologia , Infecções por Chlamydophila/imunologia , Chlamydophila pneumoniae/imunologia , Células Dendríticas/imunologia , Antígenos CD/biossíntese , Antígeno B7-1/biossíntese , Linhagem Celular , Células Dendríticas/citologia , Células Dendríticas/microbiologia , Células Epiteliais/imunologia , Antígenos HLA-DR/biossíntese , Humanos , Imunoglobulinas/biossíntese , Interferon gama/metabolismo , Interleucina-12/metabolismo , Interleucina-2/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Glicoproteínas de Membrana/biossíntese , Receptores CCR7/biossíntese , Fator de Necrose Tumoral alfa/metabolismo , Antígeno CD83RESUMO
BACKGROUND: Multiple sclerosis (MS) is an immune-mediated inflammatory disease of the central nervous system. B cells have been strongly implicated in disease pathogenesis based on clinical trials with B-cell ablation. There is a growing body of evidence linking microRNAs with regulation of the immune system. Dicer, a key enzyme involved in microRNA biogenesis, is necessary for normal B-cell function. OBJECTIVE: We aimed to determine whether Dicer expression is impaired in B cells and is linked to increased expression co-stimulatory molecules in patients with MS. METHODS: B cells were separated from blood samples of MS patients and healthy subjects. Expression of Dicer and co-stimulatory molecules CD80 and CD86 was tested. The effect of Dicer modulation on CD80 and CD86 expression in B cells was studied. RESULTS: Dicer expression was decreased in B cells but not in monocytes of patients with MS compared with healthy subjects. CD80 and CD86 expression was increased on B cells of MS patients compared with healthy subjects. Inhibition of Dicer expression in B cells by small interfering RNA led to increased expression of CD80. CONCLUSION: Dicer expression is decreased and is mechanistically linked to increased expression of co-stimulatory molecule CD80 in B cells of patients with MS. This may contribute to activation of immune responses in MS.
Assuntos
Linfócitos B/imunologia , Antígeno B7-1/biossíntese , RNA Helicases DEAD-box/biossíntese , Regulação da Expressão Gênica/imunologia , Esclerose Múltipla Recidivante-Remitente/imunologia , Ribonuclease III/biossíntese , Adulto , Linfócitos B/metabolismo , Antígeno B7-1/imunologia , Antígeno B7-2/biossíntese , Antígeno B7-2/imunologia , Western Blotting , Doenças Desmielinizantes/imunologia , Feminino , Citometria de Fluxo , Humanos , Masculino , Reação em Cadeia da Polimerase em Tempo RealRESUMO
Chronic beryllium disease (CBD) is an occupational lung disorder characterized by granulomatous inflammation and the accumulation of beryllium-responsive CD4(+) T cells in the lung. These differentiated effector memory T cells secrete IL-2, IFN-γ, and TNF-α upon in vitro activation. Beryllium-responsive CD4(+) T cells in the lung are CD28 independent and have increased expression of the coinhibitory receptor, programmed death 1, resulting in Ag-specific T cells that proliferate poorly yet retain the ability to express Th1-type cytokines. To further investigate the role of coinhibitory receptors in the beryllium-induced immune response, we examined the expression of CTLA-4 in blood and bronchoalveolar lavage cells from subjects with CBD. CTLA-4 expression was elevated on CD4(+) T cells from the lungs of study subjects compared with blood. Furthermore, CTLA-4 expression was greatest in the beryllium-responsive subset of CD4(+) T cells that retained the ability to proliferate and express IL-2. Functional assays show that the induction of CTLA-4 signaling in blood cells inhibited beryllium-induced T cell proliferation while having no effect on the proliferative capacity of beryllium-responsive CD4(+) T cells in the lung. Collectively, our findings suggest a dysfunctional CTLA-4 pathway in the lung and its potential contribution to the persistent inflammatory response that characterizes CBD.
Assuntos
Beriliose/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígeno CTLA-4/imunologia , Pulmão/imunologia , Subpopulações de Linfócitos T/imunologia , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Antígeno B7-2/biossíntese , Antígeno B7-2/genética , Beriliose/sangue , Beriliose/patologia , Líquido da Lavagem Broncoalveolar/imunologia , Antígenos CD28/imunologia , Linfócitos T CD4-Positivos/patologia , Antígeno CTLA-4/biossíntese , Antígeno CTLA-4/genética , Divisão Celular , Doença Crônica , Regulação da Expressão Gênica/imunologia , Humanos , Memória Imunológica , Interferon gama/biossíntese , Interferon gama/genética , Interleucina-2/metabolismo , Pulmão/patologia , Ativação Linfocitária , Linfocinas/biossíntese , Linfocinas/genética , Modelos Imunológicos , Receptor de Morte Celular Programada 1/análise , Subpopulações de Linfócitos T/patologia , Células Th1/imunologia , Células Th1/patologiaRESUMO
Foxp3(+) T regulatory cells (Tregs) are critically important for the maintenance of immunological tolerance, immune homeostasis, and prevention of autoimmunity. Dendritic cells (DCs) are one of the major targets of Treg-mediated suppression. Some studies have suggested that Treg-mediated suppression of DC function is mediated by the interaction of CTLA-4 on Tregs with CD80/CD86 on the DCs resulting in downregulation of CD80/CD86 expression and a decrease in costimulation. We have re-examined the effects of Tregs on mouse DC function in a model in which Ag-specific, induced Tregs (iTregs) are cocultured with DCs in the absence of T effector cells. iTreg-treated DCs are markedly defective in their capacity to activate naive T cells. iTregs from CTLA-4-deficient mice failed to induce downregulation of CD80/CD86, but DCs treated with CTLA-4-deficient iTregs still exhibited impaired capacity to activate naive T cells. The iTreg-induced defect in DC function could be completely reversed by anti-IL-10, and IL-10-deficient iTregs failed to downregulate DC function. iTreg-treated DCs expressed high levels of MARCH1, an E3 ubiquitin ligase, recently found to degrade CD86 and MHC class II on the DCs and expressed lower levels of CD83, a molecule involved in neutralizing the function of MARCH1. Both the enhanced expression of MARCH1 and the decreased expression of CD83 were mediated by IL-10 produced by the iTregs. Taken together, these studies demonstrate that a major suppressive mechanism of DC function by iTregs is secondary to the effects of IL-10 on MARCH1 and CD83 expression.
Assuntos
Antígenos CD/fisiologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Regulação da Expressão Gênica/imunologia , Tolerância Imunológica/imunologia , Imunoglobulinas/fisiologia , Interleucina-10/fisiologia , Glicoproteínas de Membrana/fisiologia , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T Reguladores/imunologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Apresentação de Antígeno , Antígenos CD/biossíntese , Antígenos CD/genética , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Antígeno B7-2/biossíntese , Antígeno B7-2/genética , Linfócitos T CD4-Positivos/imunologia , Antígeno CTLA-4/deficiência , Antígeno CTLA-4/fisiologia , Separação Celular , Células Cultivadas , Técnicas de Cocultura , Proteínas de Ligação a DNA/deficiência , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/imunologia , Imunoglobulinas/biossíntese , Imunoglobulinas/genética , Interleucina-10/antagonistas & inibidores , Interleucina-10/deficiência , Interleucina-10/metabolismo , Ativação Linfocitária , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/biossíntese , Linfócitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética , Antígeno CD83RESUMO
Understanding intrathymic T cell differentiation has been greatly aided by the development of various reductionist in vitro models that mimic certain steps/microenvironments of this complex process. Most models focused on the faithful in vitro restoration of T cell differentiation and selection. In contrast, suitable in vitro models emulating the developmental pathways of the two major thymic epithelial cell lineages--cortical thymic epithelial cells and medullary thymic epithelial cells (mTECs)--are yet to be developed. In this regard, lack of an in vitro model mimicking the developmental biology of the mTEC lineage has hampered the molecular analysis of the so-called "promiscuous expression" of tissue-restricted genes, a key property of terminally differentiated mTECs. Based on the close biological relationship between the skin and thymus epithelial cell compartments, we adapted a three-dimensional organotypic coculture model, originally developed to provide a bona fide in vitro dermal equivalent, for the culture of isolated mTECs. This three-dimensional model preserves key features of mTECs: proliferation and terminal differentiation of CD80(lo), Aire(-) mTECs into CD80(hi), Aire(+) mTECs; responsiveness to RANKL; and sustained expression of FoxN1, Aire, and tissue-restricted genes in CD80(hi) mTECs. This in vitro culture model should facilitate the identification of molecular components and pathways involved in mTEC differentiation in general and in promiscuous gene expression in particular.
Assuntos
Técnicas de Cultura de Células/métodos , Células Epiteliais/citologia , Regulação da Expressão Gênica , Linfopoese , Técnicas de Cultura de Órgãos/métodos , Linfócitos T/citologia , Timócitos/citologia , Timo/citologia , Animais , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Técnicas de Cultura de Células/instrumentação , Divisão Celular , Linhagem da Célula , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/imunologia , Técnicas de Cocultura , Derme/citologia , Células Epiteliais/efeitos dos fármacos , Feminino , Fator 7 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos da radiação , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Humanos , Linfopoese/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Técnicas de Cultura de Órgãos/instrumentação , Especificidade de Órgãos , Gravidez , Ligante RANK/farmacologia , Linfócitos T/efeitos dos fármacos , Timócitos/efeitos dos fármacos , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteína AIRERESUMO
Mast cells (MCs) are immune cells residing in tissues where pathogens are first encountered. It has been indicated that MCs might also be involved in setting the outcome of T-cell responses. However, little is known about the capacity of human MCs to express MHC class II and/or to capture and present antigens to CD4(+) T cells. To study the T-cell stimulatory potential of human MCs, CD34(+) stem cell derived MCs were generated. These cells expressed HLA-DR when stimulated with IFN-γ, and, importantly, presented peptide and protein for activation of antigen-specific CD4(+) T cells. The interplay between MC and T cell led to increased HLA-DR expression on MCs. MCs were present in close proximity to T cells in tonsil and expressed HLA-DR and CD80, indicating their ability to present antigens to CD4(+) T cells in T-cell areas of human LNs. Our data show that MCs can present native antigens to human CD4(+) T cells and that HLA-DR expressing MCs are present in tonsil tissue, indicating that human MCs can directly activate T cells and provide a rationale to study the potential of MCs to prime and/or skew human T-cell responses.