Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
1.
Nat Immunol ; 24(6): 915-924, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37081147

RESUMO

Immune cell locomotion is associated with amoeboid migration, a flexible mode of movement, which depends on rapid cycles of actin polymerization and actomyosin contraction1. Many immune cells do not necessarily require integrins, the major family of adhesion receptors in mammals, to move productively through three-dimensional tissue spaces2,3. Instead, they can use alternative strategies to transmit their actin-driven forces to the substrate, explaining their migratory adaptation to changing external environments4-6. However, whether these generalized concepts apply to all immune cells is unclear. Here, we show that the movement of mast cells (immune cells with important roles during allergy and anaphylaxis) differs fundamentally from the widely applied paradigm of interstitial immune cell migration. We identify a crucial role for integrin-dependent adhesion in controlling mast cell movement and localization to anatomical niches rich in KIT ligand, the major mast cell growth and survival factor. Our findings show that substrate-dependent haptokinesis is an important mechanism for the tissue organization of resident immune cells.


Assuntos
Actinas , Integrinas , Animais , Integrinas/metabolismo , Actinas/metabolismo , Mastócitos/metabolismo , Movimento Celular , Leucócitos/metabolismo , Adesão Celular , Mamíferos/metabolismo
2.
Immunity ; 55(12): 2285-2299.e7, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36272416

RESUMO

Intravascular neutrophils and platelets collaborate in maintaining host integrity, but their interaction can also trigger thrombotic complications. We report here that cooperation between neutrophil and platelet lineages extends to the earliest stages of platelet formation by megakaryocytes in the bone marrow. Using intravital microscopy, we show that neutrophils "plucked" intravascular megakaryocyte extensions, termed proplatelets, to control platelet production. Following CXCR4-CXCL12-dependent migration towards perisinusoidal megakaryocytes, plucking neutrophils actively pulled on proplatelets and triggered myosin light chain and extracellular-signal-regulated kinase activation through reactive oxygen species. By these mechanisms, neutrophils accelerate proplatelet growth and facilitate continuous release of platelets in steady state. Following myocardial infarction, plucking neutrophils drove excessive release of young, reticulated platelets and boosted the risk of recurrent ischemia. Ablation of neutrophil plucking normalized thrombopoiesis and reduced recurrent thrombosis after myocardial infarction and thrombus burden in venous thrombosis. We establish neutrophil plucking as a target to reduce thromboischemic events.


Assuntos
Doenças Cardiovasculares , Infarto do Miocárdio , Trombose , Humanos , Megacariócitos , Trombopoese , Neutrófilos , Plaquetas/fisiologia
3.
Immunity ; 54(11): 2497-2513.e9, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34562377

RESUMO

Innate-like T cell populations expressing conserved TCRs play critical roles in immunity through diverse developmentally acquired effector functions. Focusing on the prototypical lineage of invariant natural killer T (iNKT) cells, we sought to dissect the mechanisms and timing of fate decisions and functional effector differentiation. Utilizing induced expression of the semi-invariant NKT cell TCR on double positive thymocytes, an initially highly synchronous wave of iNKT cell development was triggered by brief homogeneous TCR signaling. After reaching a uniform progenitor state characterized by IL-4 production potential and proliferation, effector subsets emerged simultaneously, but then diverged toward different fates. While NKT17 specification was quickly completed, NKT1 cells slowly differentiated and expanded. NKT2 cells resembled maturing progenitors, which gradually diminished in numbers. Thus, iNKT subset diversification occurs in dividing progenitor cells without acute TCR input but utilizes multiple active cytokine signaling pathways. These data imply a two-step model of iNKT effector differentiation.


Assuntos
Citocinas/metabolismo , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Biomarcadores , Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia
4.
Nature ; 627(8003): 407-415, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38383779

RESUMO

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Assuntos
Aquaporina 4 , Autoanticorpos , Autoantígenos , Linfócitos B , Tolerância Imunológica , Neuromielite Óptica , Animais , Humanos , Camundongos , Proteína AIRE , Aquaporina 4/deficiência , Aquaporina 4/genética , Aquaporina 4/imunologia , Aquaporina 4/metabolismo , Autoanticorpos/imunologia , Autoantígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD40/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Neuromielite Óptica/imunologia , Neuromielite Óptica/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo , Transcriptoma
5.
Nat Immunol ; 18(1): 74-85, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27893700

RESUMO

The cellular sources of interleukin 6 (IL-6) that are relevant for differentiation of the TH17 subset of helper T cells remain unclear. Here we used a novel strategy for the conditional deletion of distinct IL-6-producing cell types to show that dendritic cells (DCs) positive for the signaling regulator Sirpα were essential for the generation of pathogenic TH17 cells. Using their IL-6 receptor α-chain (IL-6Rα), Sirpα+ DCs trans-presented IL-6 to T cells during the process of cognate interaction. While ambient IL-6 was sufficient to suppress the induction of expression of the transcription factor Foxp3 in T cells, trans-presentation of IL-6 by DC-bound IL-6Rα (called 'IL-6 cluster signaling' here) was needed to prevent premature induction of interferon-γ (IFN-γ) expression in T cells and to generate pathogenic TH17 cells in vivo. Our findings should guide therapeutic approaches for the treatment of TH17-cell-mediated autoimmune diseases.


Assuntos
Sistema Nervoso Central/imunologia , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Subunidade alfa de Receptor de Interleucina-6/genética , Interleucina-6/metabolismo , Células Th17/imunologia , Animais , Autoimunidade , Diferenciação Celular , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/imunologia , Fragmentos de Peptídeos/imunologia , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
6.
Nat Methods ; 21(4): 584-596, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38409225

RESUMO

Although long noncoding RNAs (lncRNAs) dominate the transcriptome, their functions are largely unexplored. The extensive overlap of lncRNAs with coding and regulatory sequences restricts their systematic interrogation by DNA-directed perturbation. Here we developed genome-scale lncRNA transcriptome screening using Cas13d/CasRx. We show that RNA targeting overcomes limitations inherent to other screening methods, thereby considerably expanding the explorable space of the lncRNAome. By evolving the screening system toward pan-cancer applicability, it supports molecular and phenotypic data integration to contextualize screening hits or infer lncRNA function. We thereby addressed challenges posed by the enormous transcriptome size and tissue specificity through a size-reduced multiplexed gRNA library termed Albarossa, targeting 24,171 lncRNA genes. Its rational design incorporates target prioritization based on expression, evolutionary conservation and tissue specificity, thereby reconciling high discovery power and pan-cancer representation with scalable experimental throughput. Applied across entities, the screening platform identified numerous context-specific and common essential lncRNAs. Our work sets the stage for systematic exploration of lncRNA biology in health and disease.


Assuntos
Neoplasias , RNA Longo não Codificante , Humanos , Perfilação da Expressão Gênica , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Guia de Sistemas CRISPR-Cas , Transcriptoma , Neoplasias/genética , Neoplasias/metabolismo
7.
Nat Immunol ; 15(11): 1079-89, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25282160

RESUMO

Humoral autoimmunity paralleled by the accumulation of follicular helper T cells (T(FH) cells) is linked to mutation of the gene encoding the RNA-binding protein roquin-1. Here we found that T cells lacking roquin caused pathology in the lung and accumulated as cells of the T(H)17 subset of helper T cells in the lungs. Roquin inhibited T(H)17 cell differentiation and acted together with the endoribonuclease regnase-1 to repress target mRNA encoding the T(H)17 cell-promoting factors IL-6, ICOS, c-Rel, IRF4, IκBNS and IκBζ. This cooperation required binding of RNA by roquin and the nuclease activity of regnase-1. Upon recognition of antigen by the T cell antigen receptor (TCR), roquin and regnase-1 proteins were cleaved by the paracaspase MALT1. Thus, this pathway acts as a 'rheostat' by translating TCR signal strength via graded inactivation of post-transcriptional repressors and differential derepression of targets to enhance T(H)17 differentiation.


Assuntos
Caspases/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Ribonucleases/metabolismo , Células Th17/citologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Genes rel/genética , Células HEK293 , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Fatores Reguladores de Interferon/genética , Interleucina-6/genética , Peptídeos e Proteínas de Sinalização Intracelular , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , Proteínas Nucleares/genética , Proteínas/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Alinhamento de Sequência , Células Th17/imunologia , Ubiquitina-Proteína Ligases/genética
8.
Immunity ; 45(4): 761-773, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27692612

RESUMO

Imiquimod is a small-molecule ligand of Toll-like receptor-7 (TLR7) that is licensed for the treatment of viral infections and cancers of the skin. Imiquimod has TLR7-independent activities that are mechanistically unexplained, including NLRP3 inflammasome activation in myeloid cells and apoptosis induction in cancer cells. We investigated the mechanism of inflammasome activation by imiquimod and the related molecule CL097 and determined that K+ efflux was dispensable for NLRP3 activation by these compounds. Imiquimod and CL097 inhibited the quinone oxidoreductases NQO2 and mitochondrial Complex I. This induced a burst of reactive oxygen species (ROS) and thiol oxidation, and led to NLRP3 activation via NEK7, a recently identified component of this inflammasome. Metabolic consequences of Complex I inhibition and endolysosomal effects of imiquimod might also contribute to NLRP3 activation. Our results reveal a K+ efflux-independent mechanism for NLRP3 activation and identify targets of imiquimod that might be clinically relevant.


Assuntos
Inflamassomos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Potássio/metabolismo , RNA Nuclear Pequeno/farmacologia , Animais , Complexo I de Transporte de Elétrons/metabolismo , Camundongos , Quinases Relacionadas a NIMA/metabolismo , Quinona Redutases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor 7 Toll-Like/metabolismo
10.
Immunity ; 41(5): 722-36, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25464853

RESUMO

Regulatory T (Treg) cells maintain immune homeostasis and prevent inflammatory and autoimmune responses. During development, thymocytes bearing a moderately self-reactive T cell receptor (TCR) can be selected to become Treg cells. Several observations suggest that also in the periphery mature Treg cells continuously receive self-reactive TCR signals. However, the importance of this inherent autoreactivity for Treg cell biology remains poorly defined. To address this open question, we genetically ablated the TCR of mature Treg cells in vivo. These experiments revealed that TCR-induced Treg lineage-defining Foxp3 expression and gene hypomethylation were uncoupled from TCR input in mature Treg cells. However, Treg cell homeostasis, cell-type-specific gene expression and suppressive function critically depend on continuous triggering of their TCR.


Assuntos
Autoimunidade/imunologia , Fatores de Transcrição Forkhead/biossíntese , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Metilação de DNA/imunologia , Proteínas de Ligação a DNA/genética , Fatores de Transcrição Forkhead/genética , Inflamação/imunologia , Fatores Reguladores de Interferon/biossíntese , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/metabolismo , Timócitos/citologia
11.
Cell Mol Life Sci ; 80(1): 12, 2022 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-36534167

RESUMO

Targeting KRAS downstream signaling remains an important therapeutic approach in pancreatic cancer. We used primary pancreatic ductal epithelial cells and mouse models allowing the conditional expression of oncogenic KrasG12D, to investigate KRAS signaling integrators. We observed that the AP1 family member FRA1 is tightly linked to the KRAS signal and expressed in pre-malignant lesions and the basal-like subtype of pancreatic cancer. However, genetic-loss-of-function experiments revealed that FRA1 is dispensable for KrasG12D-induced pancreatic cancer development in mice. Using FRA1 gain- and loss-of-function models in an unbiased drug screen, we observed that FRA1 is a modulator of the responsiveness of pancreatic cancer to inhibitors of the RAF-MEK-ERK cascade. Mechanistically, context-dependent FRA1-associated adaptive rewiring of oncogenic ERK signaling was observed and correlated with sensitivity to inhibitors of canonical KRAS signaling. Furthermore, pharmacological-induced degradation of FRA1 synergizes with MEK inhibitors. Our studies establish FRA1 as a part of the molecular machinery controlling sensitivity to MAPK cascade inhibition allowing the development of mechanism-based therapies.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Proteínas Proto-Oncogênicas c-fos , Animais , Camundongos , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras) , Proteínas Proto-Oncogênicas c-fos/metabolismo , Neoplasias Pancreáticas
12.
Immunity ; 38(4): 655-68, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23583643

RESUMO

The Roquin-1 protein binds to messenger RNAs (mRNAs) and regulates gene expression posttranscriptionally. A single point mutation in Roquin-1, but not gene ablation, increases follicular helper T (Tfh) cell numbers and causes lupus-like autoimmune disease in mice. In T cells, we did not identify a unique role for the much lower expressed paralog Roquin-2. However, combined ablation of both genes induced accumulation of T cells with an effector and follicular helper phenotype. We showed that Roquin-1 and Roquin-2 proteins redundantly repressed the mRNA of inducible costimulator (Icos) and identified the Ox40 costimulatory receptor as another shared mRNA target. Combined acute deletion increased Ox40 signaling, as well as Irf4 expression, and imposed Tfh differentiation on CD4(+) T cells. These data imply that both proteins maintain tolerance by preventing inappropriate T cell activation and Tfh cell differentiation, and that Roquin-2 compensates in the absence of Roquin-1, but not in the presence of its mutated form.


Assuntos
Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , RNA Mensageiro/metabolismo , Receptores OX40/metabolismo , Proteínas Repressoras/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antígenos CD4/metabolismo , Diferenciação Celular/genética , Células HEK293 , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Ligação Proteica , Receptores OX40/genética , Proteínas Repressoras/genética , Ubiquitina-Proteína Ligases/genética
13.
Nat Immunol ; 10(6): 647-54, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19412180

RESUMO

By genetically ablating IkappaB kinase (IKK)-mediated activation of the transcription factor NF-kappaB in the B cell lineage and by analyzing a mouse mutant in which immunoglobulin lambda-chain-positive B cells are generated in the absence of rearrangements in the locus encoding immunoglobulin kappa-chain, we define here two distinct, consecutive phases of early B cell development that differ in their dependence on IKK-mediated NF-kappaB signaling. During the first phase, in which NF-kappaB signaling is dispensable, predominantly kappa-chain-positive B cells are generated, which undergo efficient receptor editing. In the second phase, predominantly lambda-chain-positive B cells are generated whose development is ontogenetically timed to occur after rearrangements of the locus encoding kappa-chain. This second phase of development is dependent on NF-kappaB signals, which can be substituted by transgenic expression of the prosurvival factor Bcl-2.


Assuntos
Linfócitos B/citologia , Cadeias kappa de Imunoglobulina/metabolismo , Cadeias lambda de Imunoglobulina/metabolismo , NF-kappa B/metabolismo , Animais , Linfócitos B/metabolismo , Diferenciação Celular , Quinase I-kappa B/genética , Cadeias kappa de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2 , Transdução de Sinais
14.
Blood ; 133(24): 2597-2609, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-30962205

RESUMO

CD30 is expressed on a variety of B-cell lymphomas, such as Hodgkin lymphoma, primary effusion lymphoma, and a diffuse large B-cell lymphoma subgroup. In normal tissues, CD30 is expressed on some activated B and T lymphocytes. However, the physiological function of CD30 signaling and its contribution to the generation of CD30+ lymphomas are still poorly understood. To gain a better understanding of CD30 signaling in B cells, we studied the expression of CD30 in different murine B-cell populations. We show that B1 cells expressed higher levels of CD30 than B2 cells and that CD30 was upregulated in IRF4+ plasmablasts (PBs). Furthermore, we generated and analyzed mice expressing a constitutively active CD30 receptor in B lymphocytes. These mice displayed an increase in B1 cells in the peritoneal cavity (PerC) and secondary lymphoid organs as well as increased numbers of plasma cells (PCs). TI-2 immunization resulted in a further expansion of B1 cells and PCs. We provide evidence that the expanded B1 population in the spleen included a fraction of PBs. CD30 signals seemed to enhance PC differentiation by increasing activation of NF-κB and promoting higher levels of phosphorylated STAT3 and STAT6 and nuclear IRF4. In addition, chronic CD30 signaling led to B-cell lymphomagenesis in aged mice. These lymphomas were localized in the spleen and PerC and had a B1-like/plasmablastic phenotype. We conclude that our mouse model mirrors chronic B-cell activation with increased numbers of CD30+ lymphocytes and provides experimental proof that chronic CD30 signaling increases the risk of B-cell lymphomagenesis.


Assuntos
Linfócitos B/imunologia , Linfócitos B/patologia , Transformação Celular Neoplásica/patologia , Antígeno Ki-1/imunologia , Linfoma de Células B/metabolismo , Animais , Antígeno Ki-1/metabolismo , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Camundongos , Camundongos Transgênicos , Plasmócitos/metabolismo , Plasmócitos/patologia , Células Precursoras de Linfócitos B/metabolismo , Células Precursoras de Linfócitos B/patologia , Transdução de Sinais/fisiologia
15.
J Immunol ; 198(7): 2747-2759, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28188245

RESUMO

NKT cells represent a small subset of glycolipid-recognizing T cells that are heavily implicated in human allergic, autoimmune, and malignant diseases. In the thymus, precursor cells recognize self-glycolipids by virtue of their semi-invariant TCR, which triggers NKT cell lineage commitment and maturation. During their development, NKT cells are polarized into the NKT1, NKT2, and NKT17 subsets, defined through their cytokine-secretion patterns and the expression of key transcription factors. However, we have largely ignored how the differentiation into the NKT cell subsets is regulated. In this article, we describe the mRNA-binding Roquin-1 and -2 proteins as central regulators of murine NKT cell fate decisions. In the thymus, T cell-specific ablation of the Roquin paralogs leads to a dramatic expansion of NKT17 cells, whereas peripheral mature NKT cells are essentially absent. Roquin-1/2-deficient NKT17 cells show exaggerated lineage-specific expression of nearly all NKT17-defining proteins tested. We show through mixed bone marrow chimera experiments that NKT17 polarization is mediated through cell-intrinsic mechanisms early during NKT cell development. In contrast, the loss of peripheral NKT cells is due to cell-extrinsic factors. Surprisingly, Roquin paralog-deficient NKT cells are, in striking contrast to conventional T cells, compromised in their ability to secrete cytokines. Altogether, we show that Roquin paralogs regulate the development and function of NKT cell subsets in the thymus and periphery.


Assuntos
Diferenciação Celular/imunologia , Células T Matadoras Naturais/imunologia , Subpopulações de Linfócitos T/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Citometria de Fluxo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
J Immunol ; 199(7): 2356-2365, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28842469

RESUMO

Maintaining immune tolerance requires the production of Foxp3-expressing regulatory T (Treg) cells in the thymus. Activation of NF-κB transcription factors is critically required for Treg cell development, partly via initiating Foxp3 expression. NF-κB activation is controlled by a negative feedback regulation through the ubiquitin editing enzyme A20, which reduces proinflammatory signaling in myeloid cells and B cells. In naive CD4+ T cells, A20 prevents kinase RIPK3-dependent necroptosis. Using mice deficient for A20 in T lineage cells, we show that thymic and peripheral Treg cell compartments are quantitatively enlarged because of a cell-intrinsic developmental advantage of A20-deficient thymic Treg differentiation. A20-deficient thymic Treg cells exhibit reduced dependence on IL-2 but unchanged rates of proliferation and apoptosis. Activation of the NF-κB transcription factor RelA was enhanced, whereas nuclear translocation of c-Rel was decreased in A20-deficient thymic Treg cells. Furthermore, we found that the increase in Treg cells in T cell-specific A20-deficient mice was already observed in CD4+ single-positive CD25+ GITR+ Foxp3- thymic Treg cell progenitors. Treg cell precursors expressed high levels of the tumor necrosis factor receptor superfamily molecule GITR, whose stimulation is closely linked to thymic Treg cell development. A20-deficient Treg cells efficiently suppressed effector T cell-mediated graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, suggesting normal suppressive function. Holding thymic production of natural Treg cells in check, A20 thus integrates Treg cell activity and increased effector T cell survival into an efficient CD4+ T cell response.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T Reguladores/fisiologia , Timo/citologia , Timo/fisiologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose , Diferenciação Celular , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Proteína Relacionada a TNFR Induzida por Glucocorticoide/genética , Doença Enxerto-Hospedeiro/prevenção & controle , Interleucina-2/imunologia , Ativação Linfocitária , Camundongos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-rel/genética , Transdução de Sinais , Transplante de Células-Tronco , Timo/imunologia , Fator de Transcrição RelA/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/deficiência
17.
Proc Natl Acad Sci U S A ; 113(18): 5065-70, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27099294

RESUMO

Although canonical NF-κB signaling is crucial to generate a normal mature B-cell compartment, its role in the persistence of resting mature B cells is controversial. To resolve this conflict, we ablated NF-κB essential modulator (NEMO) and IκB kinase 2 (IKK2), two essential mediators of the canonical pathway, either early on in B-cell development or specifically in mature B cells. Early ablation severely inhibited the generation of all mature B-cell subsets, but follicular B-cell numbers could be largely rescued by ectopic expression of B-cell lymphoma 2 (Bcl2), despite a persisting block at the transitional stage. Marginal zone (MZ) B and B1 cells were not rescued, indicating a possible role of canonical NF-κB signals beyond the control of cell survival in these subsets. When canonical NF-κB signaling was ablated specifically in mature B cells, the differentiation and/or persistence of MZ B cells was still abrogated, but follicular B-cell numbers were only mildly affected. However, the mutant cells exhibited increased turnover as well as functional deficiencies upon activation, suggesting that canonical NF-κB signals contribute to their long-term persistence and functional fitness.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Sobrevivência Celular/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL
18.
PLoS Biol ; 12(1): e1001762, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24453940

RESUMO

Mast cells are implicated in the pathogenesis of inflammatory and autoimmune diseases. However, this notion based on studies in mast cell-deficient mice is controversial. We therefore established an in vivo model for hyperactive mast cells by specifically ablating the NF-κB negative feedback regulator A20. While A20 deficiency did not affect mast cell degranulation, it resulted in amplified pro-inflammatory responses downstream of IgE/FcεRI, TLRs, IL-1R, and IL-33R. As a consequence house dust mite- and IL-33-driven lung inflammation, late phase cutaneous anaphylaxis, and collagen-induced arthritis were aggravated, in contrast to experimental autoimmune encephalomyelitis and immediate anaphylaxis. Our results provide in vivo evidence that hyperactive mast cells can exacerbate inflammatory disorders and define diseases that might benefit from therapeutic intervention with mast cell function.


Assuntos
Anafilaxia/imunologia , Artrite Experimental/imunologia , Proteínas de Ligação a DNA/deficiência , Encefalomielite Autoimune Experimental/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Mastócitos/imunologia , Ubiquitina-Proteína Ligases/deficiência , Anafilaxia/induzido quimicamente , Anafilaxia/metabolismo , Anafilaxia/patologia , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Colágeno Tipo II/administração & dosagem , Cisteína Endopeptidases , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Dinitrofenóis/administração & dosagem , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Expressão Gênica , Imunoglobulina E/genética , Imunoglobulina E/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/genética , Interleucinas/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Transgênicos , Glicoproteína Mielina-Oligodendrócito/administração & dosagem , NF-kappa B/genética , NF-kappa B/imunologia , Fragmentos de Peptídeos/administração & dosagem , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Pyroglyphidae/imunologia , Receptores de IgE/genética , Receptores de IgE/imunologia , Receptores de Interleucina/genética , Receptores de Interleucina/imunologia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Soroalbumina Bovina/administração & dosagem , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
19.
Mol Cell Proteomics ; 14(11): 2947-60, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26311899

RESUMO

Characterization of tumors at the molecular level has improved our knowledge of cancer causation and progression. Proteomic analysis of their signaling pathways promises to enhance our understanding of cancer aberrations at the functional level, but this requires accurate and robust tools. Here, we develop a state of the art quantitative mass spectrometric pipeline to characterize formalin-fixed paraffin-embedded tissues of patients with closely related subtypes of diffuse large B-cell lymphoma. We combined a super-SILAC approach with label-free quantification (hybrid LFQ) to address situations where the protein is absent in the super-SILAC standard but present in the patient samples. Shotgun proteomic analysis on a quadrupole Orbitrap quantified almost 9,000 tumor proteins in 20 patients. The quantitative accuracy of our approach allowed the segregation of diffuse large B-cell lymphoma patients according to their cell of origin using both their global protein expression patterns and the 55-protein signature obtained previously from patient-derived cell lines (Deeb, S. J., D'Souza, R. C., Cox, J., Schmidt-Supprian, M., and Mann, M. (2012) Mol. Cell. Proteomics 11, 77-89). Expression levels of individual segregation-driving proteins as well as categories such as extracellular matrix proteins behaved consistently with known trends between the subtypes. We used machine learning (support vector machines) to extract candidate proteins with the highest segregating power. A panel of four proteins (PALD1, MME, TNFAIP8, and TBC1D4) is predicted to classify patients with low error rates. Highly ranked proteins from the support vector analysis revealed differential expression of core signaling molecules between the subtypes, elucidating aspects of their pathobiology.


Assuntos
Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Linfoma Difuso de Grandes Células B/genética , Aprendizado de Máquina , Proteínas de Neoplasias/genética , Proteoma/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Formaldeído , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Marcação por Isótopo/métodos , Linfoma Difuso de Grandes Células B/diagnóstico , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Proteínas de Neoplasias/metabolismo , Neprilisina/genética , Neprilisina/metabolismo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Análise de Componente Principal , Proteoma/metabolismo , Proteômica/métodos , Transdução de Sinais , Inclusão do Tecido , Fixação de Tecidos
20.
Eur J Immunol ; 45(6): 1614-20, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25787118

RESUMO

Replication-deficient recombinant adenoviruses are potent vectors for the efficient transient expression of exogenous genes in resting immune cells. However, most leukocytes are refractory to efficient adenoviral transduction as they lack expression of the coxsackie/adenovirus receptor (CAR). To circumvent this obstacle, we generated the R26/CAG-CARΔ1(StopF) (where R26 is ROSA26 and CAG is CMV early enhancer/chicken ß actin promoter) knock-in mouse line. This strain allows monitoring of in situ Cre recombinase activity through expression of CARΔ1. Simultaneously, CARΔ1 expression permits selective and highly efficient adenoviral transduction of immune cell populations, such as mast cells or T cells, directly ex vivo in bulk cultures without prior cell purification or activation. Furthermore, we show that CARΔ1 expression dramatically improves adenoviral infection of in vitro differentiated conventional and plasmacytoid dendritic cells (DCs), basophils, mast cells, as well as Hoxb8-immortalized hematopoietic progenitor cells. This novel dual function mouse strain will hence be a valuable tool to rapidly dissect the function of specific genes in leukocyte physiology.


Assuntos
Adenoviridae/genética , Marcação de Genes , Genes Reporter , Vetores Genéticos/genética , Recombinação Homóloga , Integrases/metabolismo , Transdução Genética , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Expressão Gênica , Marcação de Genes/métodos , Humanos , Integrases/genética , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos , Camundongos Transgênicos , Células Mieloides/citologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Especificidade de Órgãos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA