Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 44(3): 553-567, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26982364

RESUMO

Intestinal epithelial cells (IECs) regulate gut immune homeostasis, and impaired epithelial responses are implicated in the pathogenesis of inflammatory bowel diseases (IBD). IEC-specific ablation of nuclear factor κB (NF-κB) essential modulator (NEMO) caused Paneth cell apoptosis and impaired antimicrobial factor expression in the ileum, as well as colonocyte apoptosis and microbiota-driven chronic inflammation in the colon. Combined RelA, c-Rel, and RelB deficiency in IECs caused Paneth cell apoptosis but not colitis, suggesting that NEMO prevents colon inflammation by NF-κB-independent functions. Inhibition of receptor-interacting protein kinase 1 (RIPK1) kinase activity or combined deficiency of Fas-associated via death domain protein (FADD) and RIPK3 prevented epithelial cell death, Paneth cell loss, and colitis development in mice with epithelial NEMO deficiency. Therefore, NEMO prevents intestinal inflammation by inhibiting RIPK1 kinase activity-mediated IEC death, suggesting that RIPK1 inhibitors could be effective in the treatment of colitis in patients with NEMO mutations and possibly in IBD.


Assuntos
Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Celulas de Paneth/fisiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Apoptose/genética , Células Cultivadas , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelB/genética
2.
Immunity ; 42(6): 1100-15, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26084025

RESUMO

Tertiary lymphoid organs (TLOs) emerge during nonresolving peripheral inflammation, but their impact on disease progression remains unknown. We have found in aged Apoe(-/-) mice that artery TLOs (ATLOs) controlled highly territorialized aorta T cell responses. ATLOs promoted T cell recruitment, primed CD4(+) T cells, generated CD4(+), CD8(+), T regulatory (Treg) effector and central memory cells, converted naive CD4(+) T cells into induced Treg cells, and presented antigen by an unusual set of dendritic cells and B cells. Meanwhile, vascular smooth muscle cell lymphotoxin ß receptors (VSMC-LTßRs) protected against atherosclerosis by maintaining structure, cellularity, and size of ATLOs though VSMC-LTßRs did not affect secondary lymphoid organs: Atherosclerosis was markedly exacerbated in Apoe(-/-)Ltbr(-/-) and to a similar extent in aged Apoe(-/-)Ltbr(fl/fl)Tagln-cre mice. These data support the conclusion that the immune system employs ATLOs to organize aorta T cell homeostasis during aging and that VSMC-LTßRs participate in atherosclerosis protection via ATLOs.


Assuntos
Envelhecimento/imunologia , Aterosclerose/imunologia , Receptor beta de Linfotoxina/metabolismo , Miócitos de Músculo Liso/fisiologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Túnica Adventícia/imunologia , Envelhecimento/genética , Animais , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Diferenciação Celular/genética , Movimento Celular/genética , Células Cultivadas , Coristoma/imunologia , Memória Imunológica , Ativação Linfocitária/genética , Tecido Linfoide/imunologia , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética
3.
J Immunol ; 203(10): 2602-2613, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31578269

RESUMO

Foxp3+ regulatory T cells are well-known immune suppressor cells in various settings. In this study, we provide evidence that knockout of the relB gene in dendritic cells (DCs) of C57BL/6 mice results in a spontaneous and systemic accumulation of Foxp3+ T regulatory T cells (Tregs) partially at the expense of microbiota-reactive Tregs. Deletion of nfkb2 does not fully recapitulate this phenotype, indicating that alternative NF-κB activation via the RelB/p52 complex is not solely responsible for Treg accumulation. Deletion of RelB in DCs further results in an impaired oral tolerance induction and a marked type 2 immune bias among accumulated Foxp3+ Tregs reminiscent of a tissue Treg signature. Tissue Tregs were fully functional, expanded independently of IL-33, and led to an almost complete Treg-dependent protection from experimental autoimmune encephalomyelitis. Thus, we provide clear evidence that RelB-dependent pathways regulate the capacity of DCs to quantitatively and qualitatively impact on Treg biology and constitute an attractive target for treatment of autoimmune diseases but may come at risk for reduced immune tolerance in the intestinal tract.


Assuntos
Autoimunidade/genética , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Linfócitos T Reguladores/imunologia , Fator de Transcrição RelB/metabolismo , Animais , Células Cultivadas , Fatores de Transcrição Forkhead/metabolismo , Técnicas de Inativação de Genes , Homeostase/imunologia , Tolerância Imunológica/imunologia , Inflamação/imunologia , Interleucina-33/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidade p52 de NF-kappa B/metabolismo , Fator de Transcrição RelB/deficiência , Fator de Transcrição RelB/genética
4.
Immunity ; 34(3): 364-74, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21419662

RESUMO

The NF-κB transcription factor regulates numerous immune responses but its contribution to interleukin-17 (IL-17) production by T cells is largely unknown. Here, we report that IL-17, but not interferon-γ (IFN-γ), production by γδ T cells required the NF-κB family members RelA and RelB as well as the lymphotoxin-ß-receptor (LTßR). In contrast, LTßR-NF-κB signaling was not involved in the differentiation of conventional αß Th17 cells. Impaired IL-17 production in RelA- or RelB-deficient T cells resulted in a diminished innate immune response to Escherichia coli infection. RelA controlled the expression of LT ligands in accessory thymocytes whereas RelB, acting downstream of LTßR, was required for the expression of RORγt and RORα4 transcription factors and the differentiation of thymic precursors into γδT17 cells. Thus, RelA and RelB within different thymocyte subpopulations cooperate in the regulation of IL-17 production by γδ T cells and contribute to the host's ability to fight bacterial infections.


Assuntos
Interleucina-17/imunologia , Linfotoxina-alfa/metabolismo , Linfócitos T/imunologia , Fator de Transcrição RelA/imunologia , Fator de Transcrição RelB/imunologia , Animais , Infecções Bacterianas/imunologia , Células Cultivadas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Timo/citologia , Timo/imunologia
5.
Eur J Immunol ; 48(6): 923-936, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29485182

RESUMO

The NF-κB transcription factor subunit RelB is important for the full activation of conventional dendritic cells (cDCs) during T-cell-dependent immune responses. Although the number of splenic DCs is greatly reduced in RelBnull mice, the cause and consequences of this deficiency are currently unknown. To circumvent the impact of the pleiotropic defects in RelBnull mice we used a reporter model for RelB expression (RelBKatushka mice) and conditionally deleted RelB in DCs (RelBCD11c-Cre mice). Thereby, we can show here that RelB is essential for the differentiation of a CD117+ CD172a+ cDC subpopulation that highly expresses RelB. Surprisingly, these DCs depend on p50 for their development and are negatively regulated by a constitutive p52 activation in absence of p100. The absence of p52/p100 had no influence on the homeostasis of CD117+ CD172a+ cDCs. RelB-dependent CD117+ CD172a+ DCs strongly induce the production of the type 2 cytokines IL-4 and IL-13, as well as GM-CSF from naïve Th cells. Consequently, mice lacking RelB in cDCs show an attenuated bronchial hyperresponsiveness with reduced eosinophil infiltration. Taken together, we have identified a new splenic RelB-dependent CD117+ CD172a+ cDC population that preferentially induces Th2 responses.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Células Dendríticas/fisiologia , Eosinófilos/imunologia , Subunidade p50 de NF-kappa B/metabolismo , Células Th2/imunologia , Fator de Transcrição RelB/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Citocinas/metabolismo , Regulação da Expressão Gênica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Imunológicos/metabolismo , Fator de Transcrição RelB/genética
6.
J Autoimmun ; 81: 56-67, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28385374

RESUMO

Medullary thymic epithelial cells (mTECs) contribute to self-tolerance by expressing and presenting peripheral tissue antigens for negative selection of autoreactive T cells and differentiation of natural regulatory T cells. The molecular control of mTEC development remains incompletely understood. We here demonstrate by TEC-specific gene manipulation in mice that the NF-κB transcription factor subunit RelB, which is activated by the alternative NF-κB pathway, regulates development of mature mTECs in a dose-dependent manner. Mice with conditional deletion of Relb lacked mature mTECs and developed spontaneous autoimmunity. In addition, the NF-κB subunits RelA and c-Rel, which are both activated by classical NF-κB signaling, were jointly required for mTEC differentiation by directly regulating the transcription of Relb. Our data reveal a crosstalk mechanism between classical and alternative NF-κB pathways that tightly controls the development of mature mTECs to ensure self-tolerance.


Assuntos
Tolerância Central/imunologia , Células Epiteliais/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Timo/imunologia , Timo/metabolismo , Animais , Autoimunidade/genética , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Epiteliais/citologia , Expressão Gênica , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo
7.
J Cell Sci ; 127(Pt 14): 3052-65, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24860143

RESUMO

NF-κB is dually involved in neurogenesis and brain pathology. Here, we addressed its role in adult axoneogenesis by generating mutations of RelA (p65) and p50 (also known as NFKB1) heterodimers of canonical NF-κB. In addition to RelA activation in astrocytes, optic nerve axonotmesis caused a hitherto unrecognized induction of RelA in growth-inhibitory oligodendrocytes. Intraretinally, RelA was induced in severed retinal ganglion cells and was also expressed in bystander Müller glia. Cell-type-specific deletion of transactivating RelA in neurons and/or macroglia stimulated axonal regeneration in a distinct and synergistic pattern. By contrast, deletion of the p50 suppressor subunit promoted spontaneous and post-injury Wallerian degeneration. Growth effects mediated by RelA deletion paralleled a downregulation of growth-inhibitory Cdh1 (officially known as FZR1) and upregulation of the endogenous Cdh1 suppressor EMI1 (officially known as FBXO5). Pro-degenerative loss of p50, however, stabilized retinal Cdh1. In vitro, RelA deletion elicited opposing pro-regenerative shifts in active nuclear and inactive cytoplasmic moieties of Cdh1 and Id2. The involvement of NF-κB and cell-cycle regulators such as Cdh1 in regenerative processes of non-replicative neurons suggests novel mechanisms by which molecular reprogramming might be executed to stimulate adult axoneogenesis and treat central nervous system (CNS) axonopathies.


Assuntos
Axônios/fisiologia , Sistema Nervoso Central/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Regeneração Nervosa/fisiologia , Células Ganglionares da Retina/fisiologia , Fator de Transcrição RelA/metabolismo , Animais , Transporte Axonal , Axônios/metabolismo , Proteínas Cdh1/metabolismo , Proteína 2 Inibidora de Diferenciação/metabolismo , Camundongos , Camundongos Transgênicos , Células Ganglionares da Retina/metabolismo , Degeneração Walleriana/metabolismo , Degeneração Walleriana/patologia
8.
Circ Res ; 114(11): 1772-87, 2014 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-24855201

RESUMO

Tertiary lymphoid organs emerge in tissues in response to nonresolving inflammation. Recent research characterized artery tertiary lymphoid organs in the aorta adventitia of aged apolipoprotein E-deficient mice. The atherosclerosis-associated lymphocyte aggregates are organized into distinct compartments, including separate T-cell areas harboring conventional, monocyte-derived, lymphoid, and plasmacytoid dendritic cells, as well as activated T-cell effectors and memory cells; B-cell follicles containing follicular dendritic cells in activated germinal centers; and peripheral niches of plasma cells. Artery tertiary lymphoid organs show marked neoangiogenesis, aberrant lymphangiogenesis, and extensive induction of high endothelial venules. Moreover, newly formed lymph node-like conduits connect the external lamina with high endothelial venules in T-cell areas and also extend into germinal centers. Mouse artery tertiary lymphoid organs recruit large numbers of naïve T cells and harbor lymphocyte subsets with opposing activities, including CD4(+) and CD8(+) effector and memory T cells, natural and induced CD4(+) regulatory T cells, and memory B cells at different stages of differentiation. These data suggest that artery tertiary lymphoid organs participate in primary immune responses and organize T- and B-cell autoimmune responses in advanced atherosclerosis. In this review, we discuss the novel concept that pro- and antiatherogenic immune responses toward unknown arterial wall-derived autoantigens may be organized by artery tertiary lymphoid organs and that disruption of the balance between pro- and antiatherogenic immune cell subsets may trigger clinically overt atherosclerosis.


Assuntos
Imunidade Adaptativa/fisiologia , Túnica Adventícia/fisiopatologia , Artérias/fisiopatologia , Aterosclerose/imunologia , Aterosclerose/fisiopatologia , Imunidade Inata/fisiologia , Tecido Linfoide/fisiopatologia , Animais , Apolipoproteínas E/deficiência , Aterosclerose/patologia , Autoimunidade/imunologia , Autoimunidade/fisiologia , Linfócitos B/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Neovascularização Patológica/fisiopatologia , Índice de Gravidade de Doença , Linfócitos T/patologia
9.
J Biol Chem ; 289(11): 7349-61, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24488495

RESUMO

The alternative nuclear factor-κB (NF-κB) pathway, mainly the RelB-p52 heterodimer, plays important roles in bone metabolism through an unknown mechanism. We have previously reported that alymphoplasia (aly/aly) mice, which lack active NF-κB-inducing kinase (NIK), show mild osteopetrosis due to the inhibition of osteoclastogenesis. p100 retains RelB in the cytoplasm and inhibits RANKL-induced osteoclastogenesis in aly/aly cells. Furthermore, the overexpression of RelB in aly/aly cells rescues RANKL-induced osteoclastogenesis by inducing p100 processing. In contrast, the overexpression of p65 in aly/aly cells has no effect. However, the overexpression of RelB fails to rescue RANKL-induced osteoclastogenesis in the presence of p100ΔGRR, which cannot be processed to p52, suggesting that p100 processing is a key step in RelB-rescued, RANKL-induced osteoclastogenesis in aly/aly cells. In this study, Cot (cancer Osaka thyroid), an MAP3K, was up-regulated by RelB overexpression. Analysis of the Cot promoter demonstrated that p65 and RelB bound to the distal NF-κB-binding site and that RelB but not p65 bound to the proximal NF-κB-binding site in the Cot promoter. The knocking down of Cot expression significantly reduced the RANKL-induced osteoclastogenesis induced by RelB overexpression. The phosphorylation of IKKα at threonine 23 and its kinase activity were indispensable for the processing of p100 and osteoclastogenesis by RelB-induced Cot. Finally, constitutively activated Akt enhanced osteoclastogenesis by RelB-induced Cot, and a dominant-negative form of Akt significantly inhibited it. Taken together, these results indicate that the overexpression of RelB restores RANKL-induced osteoclastogenesis by activation of Akt/Cot/IKKα-induced p100 processing.


Assuntos
Quinase I-kappa B/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Subunidade p52 de NF-kappa B/metabolismo , Osteoclastos/citologia , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição RelB/metabolismo , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Glutationa Transferase/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/citologia , Masculino , Camundongos , Camundongos Transgênicos , Osteogênese , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante RANK/metabolismo , Retroviridae/metabolismo , Transdução de Sinais
10.
Eur J Immunol ; 44(3): 662-72, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24242887

RESUMO

Formation of the splenic marginal zone (MZ) depends on the alternative NF-κB signaling pathway. Recently, we reported that unrestricted activation of this pathway in NF-κB2/p100-deficient (p100(-/-) ) knock-in mice alters the phenotype of MZ stroma and B cells. Here, we show that lack of the p100 inhibitor resulted in an expansion of both MZ B and peritoneal B-1 cells. However, these cells failed to generate proliferating blasts in response to T-cell-independent type 2 (TI-2) Ags, correlating with dampened IgM and absent IgG3 responses. This phenotype was in part due to increased activity of the NF-κB subunit RelB. Moreover, p100(-/-) →B6 BM chimeras were more susceptible to infection by encapsulated Streptococcus pneumoniae bacteria, pathogens that induce TI-2 responses. In contrast to the TI-2 defect, p100 deficiency did not impair immune responses to the TI-1 Ag LPS and p100(-/-) MZ B cells showed normal Ag transportation into B-cell follicles. Furthermore, p100(-/-) MZ B and B-1 cells failed to respond to TI-2 Ags in the presence of WT accessory cells. Thus, NF-κB2/p100 deficiency caused a predominant B-cell-intrinsic TI-2 defect that could largely be attributed to impaired proliferation of plasmablasts. Importantly, p100 was also necessary for efficient defense against clinically relevant TI-2 pathogens.


Assuntos
Antígenos T-Independentes/imunologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Subunidade p52 de NF-kappa B/deficiência , Animais , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Antígenos T-Independentes/metabolismo , Subpopulações de Linfócitos B/efeitos dos fármacos , Bactérias/imunologia , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Movimento Celular/genética , Citocinas/farmacologia , Predisposição Genética para Doença , Imunidade Humoral/genética , Camundongos , Camundongos Knockout , Subunidade p52 de NF-kappa B/genética , Subunidade p52 de NF-kappa B/metabolismo , Ligação Proteica , Transporte Proteico , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores Toll-Like/metabolismo , Fator de Transcrição RelB/genética , Fator de Transcrição RelB/metabolismo
11.
Immunol Cell Biol ; 93(6): 558-66, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25601276

RESUMO

The cross talk between thymocytes and the thymic epithelium is critical for T-cell development and the establishment of central tolerance. Medullary thymic epithelial cells (mTECs) are located in the thymic medulla and mediate the elimination of self-reactive thymocytes, thereby preventing the onset of autoimmunity. Previous studies identified the deubiquitinating enzyme CYLD as a critical regulator of T-cell development by activating proximal T-cell receptor signaling during the transition of double-positive to single-positive thymocytes. Here we evaluated the impact of the naturally occurring short-splice variant of the cyld gene (sCYLD) on the development and maturation of mTECs. We found that thymi of CYLD(ex7/8) mice, solely expressing sCYLD, displayed a reduced number of mature mTECs caused by a developmental block during the transition of immature to mature mTECs. Further, we could demonstrate an impaired negative selection of thymocytes in these mice. Our data demonstrate that inefficient negative selection in the thymus of CYLD(ex7/8) mice result from a defect in mTEC maturation.


Assuntos
Diferenciação Celular , Cisteína Endopeptidases/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Timo/citologia , Timo/metabolismo , Animais , Antígenos de Superfície/metabolismo , Contagem de Células , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Cisteína Endopeptidases/genética , Enzima Desubiquitinante CYLD , Feminino , Imunofenotipagem , Camundongos , Camundongos Knockout , Mutação , Fenótipo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Timócitos/imunologia , Timócitos/metabolismo , Ubiquitinação
12.
PLoS Pathog ; 8(3): e1002555, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22396644

RESUMO

Microbial pathogens have developed efficient strategies to compromise host immune responses. Cryptococcus neoformans is a facultative intracellular pathogen, recognised as the most common cause of systemic fungal infections leading to severe meningoencephalitis, mainly in immunocompromised patients. This yeast is characterized by a polysaccharide capsule, which inhibits its phagocytosis. Whereas phagocytosis escape and macrophage intracellular survival have been intensively studied, extracellular survival of this yeast and restraint of host innate immune response are still poorly understood. In this study, we have investigated whether C. neoformans affected macrophage cell viability and whether NF-κB (nuclear factor-κB), a key regulator of cell growth, apoptosis and inflammation, was involved. Using wild-type (WT) as well as mutant strains of C. neoformans for the pathogen side, and WT and mutant cell lines with altered NF-κB activity or signalling as well as primary macrophages for the host side, we show that C. neoformans manipulated NF-κB-mediated signalling in a unique way to regulate macrophage cell fate and viability. On the one hand, serotype A strains reduced macrophage proliferation in a capsule-independent fashion. This growth decrease, which required a critical dosage of NF-κB activity, was caused by cell cycle disruption and aneuploidy, relying on fungal-induced modification of expression of several cell cycle checkpoint regulators in S and G2/M phases. On the other hand, C. neoformans infection induced macrophage apoptosis in a capsule-dependent manner with a differential requirement of the classical and alternative NF-κB signalling pathways, the latter one being essential. Together, these findings shed new light on fungal strategies to subvert host response through uncoupling of NF-κB activity in pathogen-controlled apoptosis and impairment of cell cycle progression. They also provide the first demonstration of induction of aneuploidy by a fungal pathogen, which may have wider implications for human health as aneuploidy is proposed to promote tumourigenesis.


Assuntos
Apoptose/fisiologia , Pontos de Checagem do Ciclo Celular/genética , Instabilidade Cromossômica , Cryptococcus neoformans/patogenicidade , Macrófagos/microbiologia , NF-kappa B/genética , Aneuploidia , Animais , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Criptococose/imunologia , Criptococose/metabolismo , Cryptococcus neoformans/imunologia , Cryptococcus neoformans/metabolismo , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , NF-kappa B/metabolismo , Transdução de Sinais
13.
Neuroimage ; 59(1): 363-76, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-21835252

RESUMO

Traditionally, depiction of isolated CNS fiber tracts is achieved by histological post mortem studies. As a tracer-dependent strategy, the calcium analog manganese has proved valuable for in vivo imaging of CNS trajectories, particularly in rats. However, adequate protocols in mice are still rare. To take advantage of the numerous genetic mouse mutants that are available to study axonal de- and regeneration processes, a MnCl2-based protocol for high-resolution contrast-enhanced MRI (MEMRI) of the visual pathway in mice acquired on a widely used clinical 3 Tesla scanner was established. Intravitreal application of MnCl2 significantly enhanced T1-weighted contrast and signal intensity along the retino-petal projection enabling its reconstruction in a 3D mode from a maximum intensity projection (MIP) calculated dataset. In response to crush injury of the optic nerve, axonal transport of MnCl2 was diminished and completely blocked proximal and distal to the lesion site, respectively. Conditions of Wallerian degeneration after acute optic nerve injury accelerated Mn2+-enhanced signal fading in axotomized projection areas between 12 and 24 h post-injury. In long-term regeneration studies 12 months after optic nerve injury, the MRI protocol proved highly sensitive and discriminated animals with rare spontaneous axonal regrowth from non-regenerating specimens. Also, structural MRI aspects shared high correlation with histological results in identical animals. Moreover, in a model of chronic neurodegeneration in p50/NF-κB-deficient mice, MnCl2-based neuron-axonal tracing supported by heat map imaging indicated neuropathy of the visual pathway due to atrophy of optic nerve fiber projections. Toxic effects of MnCl2 at MRI contrast-relevant dosages in repetitive administration protocols were ruled out by histological and optometric examinations. At higher dosages, photoreceptors, not retinal ganglion cells, turned out as most susceptible to the well-known toxicity of MnCl2. Our data accentuate in vivo MEMRI of the murine visual system as a highly specific and sensitive strategy to uncover axonal degeneration and restoration processes, even in a functional latent state. We expect MEMRI to be promising for future applications in longitudinal studies on development, aging, or regeneration of CNS projections in mouse models mimicking human CNS pathologies.


Assuntos
Cloretos , Aumento da Imagem/métodos , Imageamento por Ressonância Magnética/métodos , Compostos de Manganês , Degeneração Neural/patologia , Vias Visuais/patologia , Animais , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Compressão Nervosa , Regeneração Nervosa/fisiologia
14.
Eur J Immunol ; 41(5): 1420-34, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21469094

RESUMO

Tolerance to self-antigens expressed in peripheral organs is maintained by CD4(+) CD25(+) Foxp3(+) Treg cells, which are generated as a result of thymic selection or peripheral induction. Here, we demonstrate that steady-state migratory DCs from the skin mediated Treg conversion in draining lymph nodes of mice. These DCs displayed a partially mature MHC II(int) CD86(int) CD40(hi) CCR7(+) phenotype, used endogenous TGF-ß for conversion and showed nuclear RelB translocation. Deficiency of the alternative NF-κB signaling pathway (RelB/p52) reduced steady-state migration of DCs. These DCs transported and directly presented soluble OVA provided by s.c. implanted osmotic minipumps, as well as cell-associated epidermal OVA in transgenic K5-mOVA mice to CD4(+) OVA-specific TCR-transgenic OT-II T cells. The langerin(+) dermal DC subset, but not epidermal Langerhans cells, mediated conversion of naive OT-II×RAG-1(-/-) T cells into proliferating CD4(+) CD25(+) Foxp3(+) Tregs. Thus, our data suggest that steady-state migratory RelB(+) TGF-ß(+) langerin(+) dermal DCs mediate peripheral Treg conversion in response to epidermal antigen in skin-draining lymph nodes.


Assuntos
Células de Langerhans/imunologia , Linfonodos/imunologia , Pele/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD/análise , Antígenos de Superfície/análise , Antígenos CD4/análise , Diferenciação Celular , Movimento Celular , Imunofluorescência , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Imunofenotipagem , Cadeias alfa de Integrinas/análise , Subunidade alfa de Receptor de Interleucina-2/análise , Células de Langerhans/metabolismo , Lectinas Tipo C/análise , Linfonodos/metabolismo , Complexo Principal de Histocompatibilidade , Lectinas de Ligação a Manose/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , NF-kappa B/deficiência , NF-kappa B/imunologia , Receptores CCR7/análise , Tolerância a Antígenos Próprios , Linfócitos T Reguladores/metabolismo , Fator de Transcrição RelB/análise , Fator de Crescimento Transformador beta/metabolismo
15.
Front Immunol ; 13: 913275, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36110848

RESUMO

Activation of CD40-signaling contributes to the initiation, progression and drug resistance of B cell lymphomas. We contributed to this knowledge by showing that constitutive CD40-signaling in B cells induces B cell hyperplasia and finally B cell lymphoma development in transgenic mice. CD40 activates, among others, the non-canonical NF-ĸB signaling, which is constitutively activated in several human B cell lymphomas and is therefore presumed to contribute to lymphopathogenesis. This prompted us to study the regulatory role of the non-canonical NF-ĸB transcription factor RelB in lymphomagenesis. To this end, we crossed mice expressing a constitutively active CD40 receptor in B cells with conditional RelB-KO mice. Ablation of RelB attenuated pre-malignant B cell expansion, and resulted in an impaired survival and activation of long-term CD40-stimulated B cells. Furthermore, we found that hyperactivation of non-canonical NF-кB signaling enhances the retention of B cells in the follicles of secondary lymphoid organs. RNA-Seq-analysis revealed that several genes involved in B-cell migration, survival, proliferation and cytokine signaling govern the transcriptional differences modulated by the ablation of RelB in long-term CD40-stimulated B cells. Inactivation of RelB did not abrogate lymphoma development. However, lymphomas occurred with a lower incidence and had a longer latency period. In summary, our data suggest that RelB, although it is not strictly required for malignant transformation, accelerates the lymphomagenesis of long-term CD40-stimulated B cells by regulating genes involved in migration, survival and cytokine signaling.


Assuntos
Linfoma de Células B , Linfoma , Fator de Transcrição RelB , Animais , Linfócitos B , Antígenos CD40/genética , Citocinas , Humanos , Linfoma de Células B/genética , Camundongos , Camundongos Transgênicos , NF-kappa B , Fator de Transcrição RelB/genética
16.
Arterioscler Thromb Vasc Biol ; 30(3): 395-402, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20139367

RESUMO

OBJECTIVE: Mouse aorta smooth muscle cells (SMC) express tumor necrosis factor receptor superfamily member 1A (TNFR-1) and lymphotoxin beta-receptor (LTbetaR). Circumstantial evidence has linked the SMC LTbetaR to tertiary lymphoid organogenesis in hyperlipidemic mice. Here, we explored TNFR-1 and LTbetaR signaling in cultured SMC. METHODS AND RESULTS: TNFR-1 signaling activated the classical RelA NF-kappaB pathway, whereas LTbetaR signaling activated the classical RelA and alternative RelB NF-kappaB pathways, and both signaling pathways synergized to enhance p100 inhibitor processing to the p52 subunit of NF-kappaB. Microarrays showed that simultaneous TNFR-1/LTbetaR activation resulted in elevated mRNA encoding leukocyte homeostatic chemokines CCL2, CCL5, CXCL1, and CX3CL1. Importantly, SMC acquired features of lymphoid tissue organizers, which control tertiary lymphoid organogenesis in autoimmune diseases through hyperinduction of CCL7, CCL9, CXCL13, CCL19, CXCL16, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1. TNFR-1/LTbetaR cross-talk resulted in augmented secretion of lymphorganogenic chemokine proteins. Supernatants of TNFR-1/LTbetaR-activated SMC markedly supported migration of splenic T cells, B cells, and macrophages/dendritic cells. Experiments with ltbr(-/-) SMC indicated that LTbetaR-RelB activation was obligatory to generate the lymphoid tissue organizer phenotype. CONCLUSIONS: SMC may participate in the formation of tertiary lymphoid tissue in atherosclerosis by upregulation of lymphorganogenic chemokines involved in T-lymphocyte, B-lymphocyte, and macrophage/dendritic cell attraction.


Assuntos
Diferenciação Celular/fisiologia , Tecido Linfoide/citologia , Receptor beta de Linfotoxina/fisiologia , Miócitos de Músculo Liso/citologia , NF-kappa B/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Transdução de Sinais/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/fisiologia , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Movimento Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Tecido Linfoide/fisiologia , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Fator de Necrose Tumoral alfa/farmacologia
17.
J Exp Med ; 197(12): 1613-21, 2003 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-12810684

RESUMO

Natural killer T (NKT) cells have been implicated in diverse immune responses ranging from suppression of autoimmunity to tumor rejection. Thymus-dependent NKT cells are positively selected by the major histocompatibility complex class I-like molecule CD1d, but the molecular events downstream of CD1d are still poorly understood. Here, we show that distinct members of the Rel/nuclear factor (NF)-kappa B family of transcription factors were required in both hematopoietic and nonhematopoietic cells for normal development of thymic NKT cells. Activation of NF-kappa B via the classical I kappa B alpha-regulated pathway was required in a cell autonomous manner for the transition of NK-1.1-negative precursors that express the TCR V alpha 14-J alpha 18 chain to mature NK-1.1-positive NKT cells. The Rel/NF-kappa B family member RelB, on the other hand, had to be expressed in radiation resistant thymic stromal cells for the generation of early NK-1.1-negative NKT precursors. Moreover, NF-kappa B-inducing kinase (NIK) was required for both constitutive thymic DNA binding of RelB and the specific induction of RelB complexes in vitro. Thus, distinct Rel/NF-kappa B family members in hematopoietic and nonhematopoietic cells regulate NKT cell development with a unique requirement for NIK-mediated activation of RelB in thymic stroma.


Assuntos
Diferenciação Celular/fisiologia , Células Matadoras Naturais/fisiologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Subpopulações de Linfócitos T/fisiologia , Animais , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Interleucina-15/genética , Interleucina-15/metabolismo , Receptor beta de Linfotoxina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-rel/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Baço/citologia , Baço/imunologia , Timo/citologia , Timo/imunologia , Timo/metabolismo , Quinase Induzida por NF-kappaB
18.
Blood ; 112(3): 551-9, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18505785

RESUMO

Nuclear factor-kappaB (NF-kappaB) transcription factors regulate B-cell development and survival. However, whether they also have a role during early steps of B-cell differentiation is largely unclear. Here, we show that constitutive activation of the alternative NF-kappaB pathway in p100(-/-) knockin mice resulted in a block of early B-cell development at the transition from the pre-pro-B to the pro-B-cell stage due to enhanced RelB activity. Expression of the essential B-cell transcription factors EBF and in particular Pax5 was reduced in p100(-/-) B-cell precursors in a RelB-dependent manner, resulting in reduced mRNA levels of B lineage-specific genes. Moreover, enhanced RelB function in p100(-/-) B-cell precursors was accompanied by increased expression of B lineage-inappropriate genes, such as C/EBP alpha, correlating with a markedly increased myeloid differentiation potential of p100(-/-) progenitor B cells. Ectopic expression of Pax5 in hematopoietic progenitors restored early B-cell development in p100(-/-) bone marrow, suggesting that impaired early B lymphopoiesis in mice lacking the p100 inhibitor may be due to down-regulation of Pax5 expression. Thus, tightly controlled p100 processing and RelB activation is essential for normal B lymphopoiesis and lymphoid/myeloid lineage decision in bone marrow.


Assuntos
Linfócitos B/citologia , Linfopoese , Subunidade p52 de NF-kappa B/fisiologia , Fator de Transcrição RelB/fisiologia , Animais , Medula Óssea , Linhagem da Célula , Camundongos , Camundongos Knockout , Subunidade p52 de NF-kappa B/deficiência , Fator de Transcrição PAX5/genética , Transativadores/genética
19.
Eur J Immunol ; 38(12): 3508-19, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19003818

RESUMO

Development of NKT cells was shown to depend on lymphotoxin (LT) and IL-15 signaling pathways as well as on cytokine receptor common gamma chain. After positive selection, NKT-cell precursors transit through progressive maturation stages including proliferative expansion within the NK1.1(-) window. The transcription factors that integrate different signaling pathways into different stages of NKT-cell development are not well characterized. Here, we show that the Rel/NF-kappaB family member RelA regulates the NK1.1(-) to NK1.1(+) transition during NKT-cell development. RelA is also required for both IL-15- and IL-7-induced proliferation of CD44(hi)NK1.1(-) NKT-cell precursors. Activation of the invariant NKT-cell receptor induces both IL-15 receptor alpha and gamma chains' expression in an NF-kappaB-dependent manner, suggesting a molecular mechanism by which NF-kappaB regulates NKT-cell development. NF-kappaB also regulates TCR-induced expression of LT-alpha and LT-beta within NKT cells. In contrast to previous reports, however, we show that LT signaling is dispensable for thymic NKT-cell development but is essential for their colonization of peripheral organs such as liver.


Assuntos
Diferenciação Celular/imunologia , Interleucina-15/farmacologia , Ligases/metabolismo , NF-kappa B/metabolismo , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Receptores de Hialuronatos/imunologia , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Subunidade alfa de Receptor de Interleucina-15/metabolismo , Interleucina-7/farmacologia , Ligases/classificação , Linfotoxina-alfa/metabolismo , Camundongos , NF-kappa B/classificação , Células T Matadoras Naturais/citologia , Células T Matadoras Naturais/efeitos dos fármacos , Ligação Proteica , Subunidades Proteicas/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia
20.
Immunobiology ; 224(5): 687-696, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31200979

RESUMO

Regulatory T cells (Tregs) maintain immune homeostasis and play an important role in tissue regeneration after injury. Mutations affecting development or homeostasis of Tregs lead to immune pathologies in humans and are often fatal in mouse models. Although the pathways required for Treg development are being increasingly characterized, factors crucial for Treg homeostasis are not completely understood. Previously we have found a role for alternative NF-κB pathway in restricting T cell activation and Th17 differentiation. Here, by using the mouse model of uncontrolled alternative NF-κB signaling we identify a crucial intrinsic role of RelB signaling in regulating homeostasis and competitive fitness of Tregs. The failure of p100-/- Tregs to maintain the population of effector Tregs and efficiently suppress immune reactions results in lethal multiorgan Th1-mediated inflammation in Rag1-/- recipients. This inflammation is combined with severe lymphopenia and could be rescued by adoptive transfer of wild type Tregs. Thus in addition to its role in Th17 differentiation, RelB acts as a potent inhibitor of Treg effector functions. Our results point to RelB as a potential therapeutic target for Treg manipulation.


Assuntos
Homeostase , NF-kappa B/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Autoimunidade , Biomarcadores , Citocinas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Imunomodulação/genética , Imunofenotipagem , Ativação Linfocitária , Camundongos , Camundongos Knockout , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Fator de Transcrição RelB/metabolismo , Proteína p120 Ativadora de GTPase/genética , Proteína p120 Ativadora de GTPase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA