Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
iScience ; 26(4): 106381, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37009211

RESUMO

Small molecule IAP antagonists - SMAC mimetics (SM) - are being developed as an anticancer therapy. SM therapy was demonstrated not only to sensitize tumor cells to TNFα-mediated cell death but also to exert immunostimulatory properties. Their good safety and tolerability profile, plus promising preclinical data, warrants further investigation into their various effects within the tumor microenvironment. Using in vitro models of human tumor cells and fibroblast spheroids co-cultured with primary immune cells, we investigated the effects of SM on immune cell activation. SM treatment induces the maturation of human PBMC- and patient-derived dendritic cells (DC), and modulates cancer-associated fibroblasts towards an immune interacting phenotype. Finally, SM-induced tumor necroptosis further enhances DC activation, leading also to higher T-cell activation and infiltration into the tumor site. These results highlight the relevance of using heterotypic in vitro models to investigate the effects of targeted therapies on different components of the tumor microenvironment.

2.
J Pharmacol Exp Ther ; 384(3): 331-342, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36241203

RESUMO

Vascular endothelial growth factor (VEGF) and angiopoietin (ANG)-2 have complementary roles in angiogenesis and promote an immunosuppressive tumor microenvironment. It is anticipated that the combination of VEGF and ANG2 blockade could provide superior activity to the blockade of either pathway alone and that the addition of VEGF/ANG2 inhibition to an anti-programmed cell death protein-1 (PD-1) antibody could change the tumor microenvironment to support T-cell-mediated tumor cytotoxicity. Here, we describe the pharmacologic and antitumor activity of BI 836880, a humanized bispecific nanobody comprising two single-variable domains blocking VEGF and ANG2, and an additional module for half-life extension in vivo. BI 836880 demonstrated high affinity and selectivity for human VEGF-A and ANG2, resulting in inhibition of the downstream signaling of VEGF/ANG2 and a decrease in endothelial cell proliferation and survival. In vivo, BI 836880 exhibited significant antitumor activity in all patient-derived xenograft models tested, showing significantly greater tumor growth inhibition (TGI) than bevacizumab (VEGF inhibition) and AMG386 (ANG1/2 inhibition) in a range of models. In a Lewis lung carcinoma syngeneic tumor model, the combination of PD-1 inhibition with VEGF inhibition showed superior efficacy versus the blockade of either pathway alone. TGI was further increased with the addition of ANG2 inhibition to VEGF/PD-1 blockade. VEGF/ANG2 inhibition had a strong antiangiogenic effect. Our data suggest that the blockade of VEGF and ANG2 with BI 836880 may offer improved antitumor activity versus the blockade of either pathway alone and that combining VEGF/ANG2 inhibition with PD-1 blockade can further enhance antitumor effects. SIGNIFICANCE STATEMENT: Vascular endothelial growth factor (VEGF) and angiopoietin (ANG)-2 play key roles in angiogenesis and have an immunosuppressive effect in the tumor microenvironment. This study shows that BI 836880, a bispecific nanobody targeting VEGF and ANG2, demonstrates substantial antitumor activity in preclinical models. Combining VEGF/ANG2 inhibition with the blockade of the PD-1 pathway can further improve antitumor activity.


Assuntos
Neoplasias , Fator A de Crescimento do Endotélio Vascular , Humanos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Angiopoietina-2/metabolismo , Receptor de Morte Celular Programada 1 , Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Inibidores da Angiogênese , Neoplasias/tratamento farmacológico , Morte Celular , Angiopoietina-1 , Microambiente Tumoral
3.
Cells ; 9(4)2020 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-32235616

RESUMO

Cancer therapies based on in vivo stimulation, or on adoptive T cell transfer of Vγ9Vδ2 T cells, have been tested in the past decades but have failed to provide consistent clinical efficacy. New, promising concepts such as γδ Chimeric Antigen Receptor (CAR) -T cells and γδ T-cell engagers are currently under preclinical evaluation. Since the impact of factors, such as the relatively low abundance of γδ T cells within tumor tissue is still under investigation, it remains to be shown whether these effector T cells can provide significant efficacy against solid tumors. Here, we highlight key learnings from the natural role of Vγ9Vδ2 T cells in the elimination of host cells bearing intracellular bacterial agents and we translate these into the setting of tumor therapy. We discuss the availability and relevance of preclinical models as well as currently available tools and knowledge from a drug development perspective. Finally, we compare advantages and disadvantages of existing therapeutic concepts and propose a role for Vγ9Vδ2 T cells in immune-oncology next to Cluster of Differentiation (CD) 3 activating therapies.


Assuntos
Infecções/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/imunologia , Animais , Plasticidade Celular , Humanos , Neoplasias/patologia , Receptores de Reconhecimento de Padrão/metabolismo
4.
Nat Protoc ; 14(11): 3082-3100, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31554955

RESUMO

Blood vessels are fundamental to animal life and have critical roles in many diseases, such as stroke, myocardial infarction and diabetes. The vasculature is formed by endothelial cells that line the vessel and are covered with mural cells, specifically pericytes in smaller vessels and vascular smooth muscle cells (vSMCs) in larger-diameter vessels. Both endothelial cells and mural cells are essential for proper blood vessel function and can be derived from human pluripotent stem cells (hPSCs). Here, we describe a protocol to generate self-organizing 3D human blood vessel organoids from hPSCs that exhibit morphological, functional and molecular features of human microvasculature. These organoids are differentiated via mesoderm induction of hPSC aggregates and subsequent differentiation into endothelial networks and pericytes in a 3D collagen I-Matrigel matrix. Blood vessels form within 2-3 weeks and can be further grown in scalable suspension culture. Importantly, in vitro-differentiated human blood vessel organoids transplanted into immunocompromised mice gain access to the mouse circulation and specify into functional arteries, arterioles and veins.


Assuntos
Vasos Sanguíneos/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Diferenciação Celular , Linhagem Celular , Colágeno/química , Combinação de Medicamentos , Endotélio Vascular/citologia , Humanos , Laminina/química , Microvasos/citologia , Neovascularização Fisiológica , Pericitos/citologia , Proteoglicanas/química , Alicerces Teciduais/química
6.
Genes Dev ; 33(11-12): 669-683, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30975723

RESUMO

The transcriptional repression of alternative lineage genes is critical for cell fate commitment. Mechanisms by which locus-specific gene silencing is initiated and heritably maintained during cell division are not clearly understood. To study the maintenance of silent gene states, we investigated how the Cd4 gene is stably repressed in CD8+ T cells. Through CRISPR and shRNA screening, we identified the histone chaperone CAF-1 as a critical component for Cd4 repression. We found that the large subunit of CAF-1, Chaf1a, requires the N-terminal KER domain to associate with the histone deacetylases HDAC1/2 and the histone demethylase LSD1, enzymes that also participate in Cd4 silencing. When CAF-1 was lacking, Cd4 derepression was markedly enhanced in the absence of the de novo DNA methyltransferase Dnmt3a but not the maintenance DNA methyltransferase Dnmt1. In contrast to Dnmt1, Dnmt3a deficiency did not significantly alter levels of DNA methylation at the Cd4 locus. Instead, Dnmt3a deficiency sensitized CD8+ T cells to Cd4 derepression mediated by compromised functions of histone-modifying factors, including the enzymes associated with CAF-1. Thus, we propose that the heritable silencing of the Cd4 gene in CD8+ T cells exploits cooperative functions among the DNA methyltransferases, CAF-1, and histone-modifying enzymes.


Assuntos
Antígenos CD4/genética , Fator 1 de Modelagem da Cromatina/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteína 4 de Ligação ao Retinoblastoma/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Animais , Antígenos CD4/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Chaperonas de Histonas/metabolismo , Histona Desacetilases/metabolismo , Histonas/metabolismo , Masculino , Camundongos , Domínios Proteicos
7.
Nature ; 565(7740): 505-510, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30651639

RESUMO

The increasing prevalence of diabetes has resulted in a global epidemic1. Diabetes is a major cause of blindness, kidney failure, heart attacks, stroke and amputation of lower limbs. These are often caused by changes in blood vessels, such as the expansion of the basement membrane and a loss of vascular cells2-4. Diabetes also impairs the functions of endothelial cells5 and disturbs the communication between endothelial cells and pericytes6. How dysfunction of endothelial cells and/or pericytes leads to diabetic vasculopathy remains largely unknown. Here we report the development of self-organizing three-dimensional human blood vessel organoids from pluripotent stem cells. These human blood vessel organoids contain endothelial cells and pericytes that self-assemble into capillary networks that are enveloped by a basement membrane. Human blood vessel organoids transplanted into mice form a stable, perfused vascular tree, including arteries, arterioles and venules. Exposure of blood vessel organoids to hyperglycaemia and inflammatory cytokines in vitro induces thickening of the vascular basement membrane. Human blood vessels, exposed in vivo to a diabetic milieu in mice, also mimic the microvascular changes found in patients with diabetes. DLL4 and NOTCH3 were identified as key drivers of diabetic vasculopathy in human blood vessels. Therefore, organoids derived from human stem cells faithfully recapitulate the structure and function of human blood vessels and are amenable systems for modelling and identifying the regulators of diabetic vasculopathy, a disease that affects hundreds of millions of patients worldwide.


Assuntos
Membrana Basal/patologia , Vasos Sanguíneos/patologia , Angiopatias Diabéticas/patologia , Modelos Biológicos , Organoides/patologia , Organoides/transplante , Proteínas Adaptadoras de Transdução de Sinal , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Artérias/citologia , Artérias/efeitos dos fármacos , Arteríolas/citologia , Arteríolas/efeitos dos fármacos , Membrana Basal/citologia , Membrana Basal/efeitos dos fármacos , Vasos Sanguíneos/citologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/crescimento & desenvolvimento , Proteínas de Ligação ao Cálcio , Angiopatias Diabéticas/enzimologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Hiperglicemia/complicações , Técnicas In Vitro , Mediadores da Inflamação/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Organoides/citologia , Organoides/efeitos dos fármacos , Pericitos/citologia , Pericitos/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Receptor Notch3/metabolismo , Transdução de Sinais , Vênulas/citologia , Vênulas/efeitos dos fármacos
8.
Nature ; 563(7732): 564-568, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30405245

RESUMO

Genetic regulators and environmental stimuli modulate T cell activation in autoimmunity and cancer. The enzyme co-factor tetrahydrobiopterin (BH4) is involved in the production of monoamine neurotransmitters, the generation of nitric oxide, and pain1,2. Here we uncover a link between these processes, identifying a fundamental role for BH4 in T cell biology. We find that genetic inactivation of GTP cyclohydrolase 1 (GCH1, the rate-limiting enzyme in the synthesis of BH4) and inhibition of sepiapterin reductase (the terminal enzyme in the synthetic pathway for BH4) severely impair the proliferation of mature mouse and human T cells. BH4 production in activated T cells is linked to alterations in iron metabolism and mitochondrial bioenergetics. In vivo blockade of BH4 synthesis abrogates T-cell-mediated autoimmunity and allergic inflammation, and enhancing BH4 levels through GCH1 overexpression augments responses by CD4- and CD8-expressing T cells, increasing their antitumour activity in vivo. Administration of BH4 to mice markedly reduces tumour growth and expands the population of intratumoral effector T cells. Kynurenine-a tryptophan metabolite that blocks antitumour immunity-inhibits T cell proliferation in a manner that can be rescued by BH4. Finally, we report the development of a potent SPR antagonist for possible clinical use. Our data uncover GCH1, SPR and their downstream metabolite BH4 as critical regulators of T cell biology that can be readily manipulated to either block autoimmunity or enhance anticancer immunity.


Assuntos
Doenças Autoimunes/imunologia , Biopterinas/análogos & derivados , Neoplasias/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Administração Oral , Oxirredutases do Álcool/antagonistas & inibidores , Oxirredutases do Álcool/metabolismo , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/patologia , Biopterinas/biossíntese , Biopterinas/metabolismo , Biopterinas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Coenzimas/metabolismo , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Feminino , GTP Cicloidrolase/genética , GTP Cicloidrolase/metabolismo , Humanos , Hipersensibilidade/imunologia , Ferro/metabolismo , Cinurenina/metabolismo , Cinurenina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
9.
Nat Commun ; 9(1): 1983, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29777171

RESUMO

MLL-fusions represent a large group of leukemia drivers, whose diversity originates from the vast molecular heterogeneity of C-terminal fusion partners of MLL. While studies of selected MLL-fusions have revealed critical molecular pathways, unifying mechanisms across all MLL-fusions remain poorly understood. We present the first comprehensive survey of protein-protein interactions of seven distantly related MLL-fusion proteins. Functional investigation of 128 conserved MLL-fusion-interactors identifies a specific role for the lysine methyltransferase SETD2 in MLL-leukemia. SETD2 loss causes growth arrest and differentiation of AML cells, and leads to increased DNA damage. In addition to its role in H3K36 tri-methylation, SETD2 is required to maintain high H3K79 di-methylation and MLL-AF9-binding to critical target genes, such as Hoxa9. SETD2 loss synergizes with pharmacologic inhibition of the H3K79 methyltransferase DOT1L to induce DNA damage, growth arrest, differentiation, and apoptosis. These results uncover a dependency for SETD2 during MLL-leukemogenesis, revealing a novel actionable vulnerability in this disease.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Leucemia/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Motivos de Aminoácidos , Diferenciação Celular , Linhagem Celular Tumoral , Dano ao DNA , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Humanos , Leucemia/genética , Leucemia/fisiopatologia , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Proteína de Leucina Linfoide-Mieloide/química , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Fusão Oncogênica/genética , Ligação Proteica
10.
Nat Methods ; 14(12): 1191-1197, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29039415

RESUMO

Pooled CRISPR screens are a powerful tool for assessments of gene function. However, conventional analysis is based exclusively on the relative abundance of integrated single guide RNAs (sgRNAs) between populations, which does not discern distinct phenotypes and editing outcomes generated by identical sgRNAs. Here we present CRISPR-UMI, a single-cell lineage-tracing methodology for pooled screening to account for cell heterogeneity. We generated complex sgRNA libraries with unique molecular identifiers (UMIs) that allowed for screening of clonally expanded, individually tagged cells. A proof-of-principle CRISPR-UMI negative-selection screen provided increased sensitivity and robustness compared with conventional analysis by accounting for underlying cellular and editing-outcome heterogeneity and detection of outlier clones. Furthermore, a CRISPR-UMI positive-selection screen uncovered new roadblocks in reprogramming mouse embryonic fibroblasts as pluripotent stem cells, distinguishing reprogramming frequency and speed (i.e., effect size and probability). CRISPR-UMI boosts the predictive power, sensitivity, and information content of pooled CRISPR screens.


Assuntos
Sistemas CRISPR-Cas/genética , Linhagem da Célula/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes , RNA Guia de Cinetoplastídeos , Análise de Célula Única/métodos , Animais , Células Cultivadas , Fibroblastos/citologia , Técnicas de Inativação de Genes , Vetores Genéticos , Camundongos , Células-Tronco Pluripotentes/citologia , Retroviridae/genética , Razão Sinal-Ruído
11.
Autophagy ; 9(6): 931-2, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23548947

RESUMO

During development in the thymus, each T lymphocyte is equipped with one, essentially unique, T cell receptor (TCR)-specificity. Due to its random nature, this process inevitably also leads to the emergence of potentially dangerous T lymphocytes that may recognize 'self.' Nevertheless, autoimmune tissue destruction, the cause of diseases such as multiple sclerosis and diabetes, is the exception rather than the rule. This state of immunological self-tolerance is to a large degree based upon a process called 'negative selection': prior to joining the circulating lymphocyte pool, immature T cells test their receptor on self-antigens within the thymic microenvironment, and TCR engagement at this immature stage elicits an apoptotic suicide program. We now find evidence that macroautophagy supports the tolerogenic presentation of self-antigens in the thymus.


Assuntos
Autofagia/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Células Epiteliais/citologia , Células Epiteliais/imunologia , Tolerância Imunológica/imunologia , Timo/citologia , Animais , Autoantígenos/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Camundongos , Camundongos Nus , Modelos Biológicos , Fagossomos/metabolismo
12.
J Exp Med ; 210(2): 287-300, 2013 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-23382543

RESUMO

Macroautophagy serves cellular housekeeping and metabolic functions through delivery of cytoplasmic constituents for lysosomal degradation. In addition, it may mediate the unconventional presentation of intracellular antigens to CD4(+) T cells; however, the physiological relevance of this endogenous MHC class II loading pathway remains poorly defined. Here, we characterize the role of macroautophagy in thymic epithelial cells (TECs) for negative selection. Direct presentation for clonal deletion of MHC class II-restricted thymocytes required macroautophagy for a mitochondrial version of a neo-antigen, but was autophagy-independent for a membrane-bound form. A model antigen specifically expressed in Aire(+) medullary TECs (mTECs) induced efficient deletion via direct presentation when targeted to autophagosomes, whereas interference with autophagosomal routing of this antigen through exchange of a single amino acid or ablation of an essential autophagy gene abolished direct presentation for negative selection. Furthermore, when this autophagy substrate was expressed by mTECs in high amounts, endogenous presentation and indirect presentation by DCs operated in a redundant manner, whereas macroautophagy-dependent endogenous loading was essential for clonal deletion at limiting antigen doses. Our findings suggest that macroautophagy supports central CD4(+) T cell tolerance through facilitating the direct presentation of endogenous self-antigens by mTECs.


Assuntos
Autofagia/imunologia , Tolerância Central , Timo/citologia , Timo/imunologia , Animais , Apresentação de Antígeno , Proteínas Aviárias/imunologia , Proteína C-Reativa/genética , Proteína C-Reativa/imunologia , Deleção Clonal , Columbidae , Citocromos c/imunologia , Células Epiteliais/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Imunológicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteína AIRE
13.
Eur J Cell Biol ; 91(1): 24-30, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21392839

RESUMO

Thymic epithelial cells (TECs) provide a highly specialized microenvironment for the generation of a functional and self-tolerant T cell repertoire. Much of our current view of TEC biology is derived from gain- or loss-of-function approaches, which have significantly contributed to our understanding of gene function in TEC development and T cell repertoire selection. Here, we will review transgenic and viral strategies that have been used to manipulate gene expression in TECs, highlight some of the shortcomings of particular currently available tools and provide a brief outline of our own attempts to more rapidly and/or more specifically assess gene function in TECs.


Assuntos
Tolerância Central/genética , Células Epiteliais/metabolismo , Expressão Gênica/imunologia , Biologia Molecular/métodos , Linfócitos T/metabolismo , Timo/metabolismo , Adenoviridae/genética , Animais , Diferenciação Celular , Linhagem da Célula , Microambiente Celular/genética , Microambiente Celular/imunologia , Tolerância Central/imunologia , Células Epiteliais/citologia , Células Epiteliais/imunologia , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Vetores Genéticos , Humanos , Lentivirus/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Técnicas de Cultura de Tecidos
14.
J Exp Med ; 208(9): 1749-56, 2011 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-21844204

RESUMO

The substitution of one amino acid in the Roquin protein by the sanroque mutation induces a dramatic autoimmune syndrome in mice. This is believed to occur through ectopic expression of inducible T cell co-stimulator (ICOS) and unrestrained differentiation of follicular T helper cells, which induce spontaneous germinal center reactions to self-antigens. In this study, we demonstrate that tissue-specific ablation of Roquin in T or B cells, in the entire hematopoietic system, or in epithelial cells of transplanted thymi did not cause autoimmunity. Loss of Roquin induced elevated expression of ICOS through T cell-intrinsic and -extrinsic mechanisms, which itself was not sufficient to break self-tolerance. Instead, ablation of Roquin in the hematopoietic system caused defined changes in immune homeostasis, including the expansion of macrophages, eosinophils, and T cell subsets, most dramatically CD8 effector-like T cells, through cell-autonomous and nonautonomous mechanisms. Germline Roquin deficiency led to perinatal lethality, which was partially rescued on the genetic background of an outbred strain. However, not even complete absence of Roquin resulted in overt self-reactivity, suggesting that the sanroque mutation induces autoimmunity through an as yet unknown mechanism.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Autoimunidade , Regulação da Expressão Gênica/imunologia , Imunidade Celular , Ubiquitina-Proteína Ligases/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/biossíntese , Antígenos de Diferenciação de Linfócitos T/genética , Autoantígenos/imunologia , Autoantígenos/metabolismo , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Eosinófilos/imunologia , Eosinófilos/metabolismo , Regulação da Expressão Gênica/genética , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
15.
Trends Immunol ; 32(5): 188-93, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21493141

RESUMO

Promiscuous expression of 'peripheral' tissue-restricted antigens (TRAs) by medullary thymic epithelial cells (mTECs) is essential for central tolerance. Remarkably, the expression of individual TRAs varies among mTECs and is confined to a perplexingly small number of cells. To reconcile this with the ensuing robust state of tolerance, one might envisage that mTECs serve primarily as an antigen reservoir, whereas tolerogenic recognition of TRAs would ultimately require antigen uptake and presentation by dendritic cells (DCs). Here, we survey the evidence for this 'antigen-spreading' scenario and relate it to findings that document autonomous antigen-presentation by mTECs. We suggest that DC-dependent and autonomous tolerogenic functions of mTECs operate in parallel, and the underlying mechanisms remain to be established.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Autoantígenos/imunologia , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Tolerância Imunológica , Timo/imunologia , Animais , Apresentação de Antígeno , Células Apresentadoras de Antígenos/citologia , Células Dendríticas/citologia , Células Epiteliais/citologia , Humanos , Camundongos , Linfócitos T/imunologia , Timo/anatomia & histologia
16.
Nat Immunol ; 11(6): 512-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20431619

RESUMO

Medullary thymic epithelial cells (mTECs) serve an essential function in central tolerance by expressing peripheral-tissue antigens. These antigens may be transferred to and presented by dendritic cells (DCs). Therefore, it is unclear whether mTECs, in addition to being an antigen reservoir, also serve a mandatory function as antigen-presenting cells. Here we diminished major histocompatibility complex (MHC) class II on mTECs through transgenic expression of a 'designer' microRNA specific for the MHC class II transactivator CIITA (called 'C2TA' here). This resulted in an enlarged polyclonal CD4(+) single-positive compartment and, among thymocytes specific for model antigens expressed in mTECs, enhanced selection of regulatory T cells (T(reg) cells) at the expense of deletion. Our data document an autonomous contribution of mTECs to both dominant and recessive mechanisms of CD4(+) T cell tolerance and support an avidity model of T(reg) cell development versus deletion.


Assuntos
Imunidade Adaptativa , Linfócitos T CD4-Positivos/imunologia , Células Epiteliais/imunologia , Tolerância Imunológica , Timo/imunologia , Animais , Apresentação de Antígeno , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Proteínas Nucleares , Timo/crescimento & desenvolvimento , Transativadores
17.
Curr Opin Immunol ; 21(1): 92-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19246181

RESUMO

Functional and biochemical assays indicate a substantial contribution of intracellularly derived peptides to the MHC class II 'ligandome'. Macroautophagy, a process traditionally known for its role in cellular housekeeping and adaptation to nutrient withdrawal, is an attractive candidate pathway for endogenous MHC class II loading. Work in cell culture systems, including antigen presentation assays, co-localization studies and sequencing of MHC class II bound peptides, demonstrates that substrates of autophagy can be loaded onto MHC class II. Advances in the development of mouse models to monitor or genetically disrupt macroautophagy now provide the basis for elucidating the immunological relevance of autophagy in vivo. Here, we will discuss recent findings suggesting a crucial role of macroautophagy in thymic epithelial cells for the generation of peptide/MHC class II ligands for positive selection and induction of T cell tolerance.


Assuntos
Apresentação de Antígeno , Autofagia/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Animais , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Tolerância Imunológica/genética , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/imunologia , Chaperonas Moleculares/imunologia , Chaperonas Moleculares/metabolismo , Peptídeos/imunologia , Peptídeos/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/metabolismo , Timo/citologia
18.
Cell Cycle ; 7(23): 3625-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19029805

RESUMO

During intrathymic generation of the T cell repertoire, a series of selection processes ensures that only self-MHC (Major Histocompatibility Complex) restricted and self-tolerant T cells are allowed to survive. Interactions with MHC ligands on the surface of thymic epithelial cells (TECs) play a pivotal role in the decision-making of developing thymocytes. A number of distinct cell-biological features of TECs have emerged that may predispose them to serve non-redundant functions in thymocyte "education". Thus, cortical TECs express a rather unique set of proteolytic enzymes for antigen processing in the context of positive selection, whereas medullary TECs "ectopically" express a plethora of otherwise strictly tissue-restricted antigens (TRAs), a property that obviously has evolved to make these self-antigens "visible" to developing thymocytes for negative selection. One of the latest additions to this growing list of functional adaptations of TECs is their constitutively high rate of autophagy. Recently, we have provided evidence that autophagy in TECs shuttles cytoplasmic self-antigens into the MHC class II loading pathway for positive selection of T cells and tolerance induction.


Assuntos
Autofagia/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Epiteliais/citologia , Células Epiteliais/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Tolerância Imunológica/imunologia , Células Estromais/citologia , Células Estromais/imunologia
19.
Autophagy ; 4(8): 1090-2, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18927488

RESUMO

During T cell development in the thymus, scanning of peptide/major histocompatibility (MHC) molecule complexes on the surface of thymic epithelial cells ensures that only useful (self-MHC restricted) and harmless (self-tolerant) thymocytes survive. In recent years, a number of distinct cell-biological features of thymic epithelial cells have been unraveled that may have evolved to render these cells particularly suited for T cell selection, e.g., cortical epithelial cells use unique proteolytic enzymes for the generation of MHC/peptide complexes, whereas medullary epithelial cells "promiscuously" express otherwise tissue-restricted self-antigens. We recently showed that macroautophagy in thymic epithelial cells contributes to CD4 T cell selection and is essential for the generation of a self-tolerant T cell repertoire. We propose that the unusually high constitutive levels of autophagy in thymic epithelial cells deliver endogenous proteins to MHC class II molecules for both positive and negative selection of developing thymocytes.


Assuntos
Autofagia/imunologia , Linfócitos T CD4-Positivos/imunologia , Timo/imunologia , Animais , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Células Epiteliais/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Peptídeos/imunologia , Timo/citologia
20.
Nature ; 455(7211): 396-400, 2008 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-18701890

RESUMO

Recognition of self-antigen-derived epitopes presented by major histocompatibility complex class II (MHC II) molecules on thymic epithelial cells (TECs) is critical for the generation of a functional and self-tolerant CD4 T-cell repertoire. Whereas haematopoietic antigen-presenting cells generate MHC-II-peptide complexes predominantly through the processing of endocytosed polypeptides, it remains unknown if and how TECs use unconventional pathways of antigen presentation. Here we address the role of macroautophagy, a process that has recently been shown to allow for endogenous MHC II loading, in T-cell repertoire selection in the mouse thymus. In contrast to most other tissues, TECs had a high constitutive level of autophagy. Genetic interference with autophagy specifically in TECs led to altered selection of certain MHC-II-restricted T-cell specificities and resulted in severe colitis and multi-organ inflammation. Our findings indicate that autophagy focuses the MHC-II-peptide repertoire of TECs on their intracellular milieu, which notably comprises a wide array of otherwise strictly 'tissue-specific' self antigens. In doing so, it contributes to T-cell selection and is essential for the generation of a self-tolerant T-cell repertoire.


Assuntos
Autofagia , Epitélio/imunologia , Tolerância Imunológica/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Animais , Proteína 5 Relacionada à Autofagia , Diferenciação Celular , Quimera/imunologia , Colite/genética , Colite/imunologia , Colite/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Feminino , Antígenos de Histocompatibilidade/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Receptores de Antígenos de Linfócitos T/imunologia , Células Estromais/citologia , Timo/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA