Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 34(4): 108674, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33503413

RESUMO

Naive and memory T cells are maintained in a quiescent state, yet capable of rapid response and differentiation to antigen challenge via molecular mechanisms that are not fully understood. In naive cells, the deletion of Foxo1 following thymic development results in the increased expression of multiple AP-1 family members, rendering T cells less able to respond to antigenic challenge. Similarly, in the absence of FOXO1, post-infection memory T cells exhibit the characteristics of extended activation and senescence. Age-based analysis of human peripheral T cells reveals that levels of FOXO1 and its downstream target, TCF7, are inversely related to host age, whereas the opposite is found for AP-1 factors. These characteristics of aging also correlate with the formation of T cells manifesting features of cellular senescence. Our work illustrates a role for FOXO1 in the active maintenance of stem-like properties in T cells at the timescales of acute infection and organismal life span.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteína Forkhead Box O1/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Linfócitos T CD8-Positivos/citologia , Senescência Celular/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Adulto Jovem
2.
Immunity ; 42(2): 239-251, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25692700

RESUMO

T follicular helper (Tfh) cells are essential in the induction of high-affinity, class-switched antibodies. The differentiation of Tfh cells is a multi-step process that depends upon the co-receptor ICOS and the activation of phosphoinositide-3 kinase leading to the expression of key Tfh cell genes. We report that ICOS signaling inactivates the transcription factor FOXO1, and a Foxo1 genetic deletion allowed for generation of Tfh cells with reduced dependence on ICOS ligand. Conversely, enforced nuclear localization of FOXO1 inhibited Tfh cell development even though ICOS was overexpressed. FOXO1 regulated Tfh cell differentiation through a broad program of gene expression exemplified by its negative regulation of Bcl6. Final differentiation to germinal center Tfh cells (GC-Tfh) was instead FOXO1 dependent as the Foxo1(-/-) GC-Tfh cell population was substantially reduced. We propose that ICOS signaling transiently inactivates FOXO1 to initiate a Tfh cell contingency that is completed in a FOXO1-dependent manner.


Assuntos
Diferenciação Celular/imunologia , Proteínas de Ligação a DNA/biossíntese , Fatores de Transcrição Forkhead/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Animais , Ativação Enzimática , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6 , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/imunologia
3.
Integr Biol (Camb) ; 5(1): 195-203, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23060228

RESUMO

Particle-based delivery of encapsulated antigens has great potential for improving vaccine constructs. In this study, we show that antigen-loaded, pH-sensitive hydrogel microparticles are taken up and presented by bone marrow-derived dendritic cells (BMDCs) in vitro and are taken up by dendritic cells (DCs) and monocytes in vivo. This uptake is irrespective of targeting antibodies. BMDCs in vitro and DCs in vivo also display upregulation of activation markers CD80 and CD86 when treated with microparticles, again with no difference in conjugated antibodies, even the agonistic CD40 antibody. We further show that these particles induce enhanced expansion of cytokine-producing CD8 T cells in response to challenge with ovalbumin-expressing vesicular stomatitis virus, in both an accelerated vaccination strategy using pre-loaded BMDCs and a traditional mouse immunization setting.


Assuntos
Antígenos/administração & dosagem , Antígenos/imunologia , Preparações de Ação Retardada/química , Células Dendríticas/imunologia , Hidrogéis/química , Animais , Células Cultivadas , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos C57BL , Microesferas
4.
Immunity ; 33(6): 890-904, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21167754

RESUMO

Foxo transcription factors integrate extrinsic signals to regulate cell division, differentiation and survival, and specific functions of lymphoid and myeloid cells. Here, we showed the absence of Foxo1 severely curtailed the development of Foxp3(+) regulatory T (Treg) cells and those that developed were nonfunctional in vivo. The loss of function included diminished CTLA-4 receptor expression as the Ctla4 gene was a direct target of Foxo1. T cell-specific loss of Foxo1 resulted in exocrine pancreatitis, hind limb paralysis, multiorgan lymphocyte infiltration, anti-nuclear antibodies and expanded germinal centers. Foxo-mediated control over Treg cell specification was further revealed by the inability of TGF-ß cytokine to suppress T-bet transcription factor in the absence of Foxo1, resulting in IFN-γ secretion. In addition, the absence of Foxo3 exacerbated the effects of the loss of Foxo1. Thus, Foxo transcription factors guide the contingencies of T cell differentiation and the specific functions of effector cell populations.


Assuntos
Antígenos CD/biossíntese , Fatores de Transcrição Forkhead/metabolismo , Proteínas com Domínio T/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Antígenos CD/genética , Autoimunidade/genética , Antígeno CTLA-4 , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica/imunologia , Tolerância Imunológica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Equilíbrio Th1-Th2 , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
5.
J Immunol ; 183(8): 4838-42, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19801509

RESUMO

Signals initiated through the TCR during development can result in either survival and differentiation or cell death. High affinity signals that induce death elicit a robust yet transient activation of signaling pathways, including Erk, whereas low affinity ligands, which promote survival, generate a gradual and weaker activation of the same pathways. It was recently demonstrated that Erk localizes to distinct cellular locations in response to high and low affinity ligands. Although a requirement for Erk in positive selection is well established, its role in negative selection is controversial and, thus, the importance of Erk relocalization during development is not understood. In this study, we examined the role of Erk in negative selection using mice that are genetically deficient in both Erk1 and Erk2 in T cells. Results from three different models reveal that thymocyte deletion remains intact in the absence of Erk.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Linfócitos T/imunologia , Timo/imunologia , Animais , Deleção Clonal , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Técnicas de Cultura de Órgãos , Linfócitos T/enzimologia , Timo/enzimologia
6.
Nat Methods ; 4(6): 511-6, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17486086

RESUMO

The ubiquitous nature of protein phosphorylation makes it challenging to map kinase-substrate relationships, which is a necessary step toward defining signaling network architecture. To trace the activity of individual kinases, we developed a semisynthetic reaction scheme, which results in the affinity tagging of substrates of the kinase in question. First, a kinase, engineered to use a bio-orthogonal ATPgammaS analog, catalyzes thiophosphorylation of its direct substrates. Second, alkylation of thiophosphorylated serine, threonine or tyrosine residues creates an epitope for thiophosphate ester-specific antibodies. We demonstrated the generality of semisynthetic epitope construction with 13 diverse kinases: JNK1, p38alpha MAPK, Erk1, Erk2, Akt1, PKCdelta, PKCepsilon, Cdk1/cyclinB, CK1, Cdc5, GSK3beta, Src and Abl. Application of this approach, in cells isolated from a mouse that expressed endogenous levels of an analog-specific (AS) kinase (Erk2), allowed purification of a direct Erk2 substrate.


Assuntos
Epitopos/química , Epitopos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Haptenos/química , Haptenos/metabolismo , Trifosfato de Adenosina/análogos & derivados , Sequência de Aminoácidos , Animais , Epitopos/imunologia , MAP Quinases Reguladas por Sinal Extracelular/genética , Duplicação Gênica , Haptenos/imunologia , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Marcação por Isótopo/métodos , Camundongos , Camundongos Knockout , Organotiofosfatos/química , Organotiofosfatos/metabolismo , Especificidade por Substrato
7.
Immunity ; 23(4): 431-43, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16226508

RESUMO

Activation of extracellular-signal-regulated protein kinase (Erk) is central to growth-factor-receptor-mediated signaling including that originating from the T cell antigen receptor. It integrates cytoplasmic signals to effect changes in transcription associated with differentiation, proliferation, and survival. In this report, we present an analysis of mice with targeted deletions in Erk1 and Erk2 to assess the relationship between Erk activity and cell-cycle progression, thymocyte development, and lineage commitment. These studies show that Erk is selectively retained during beta selection-driven proliferation, and yet Erk1/2 are not required to complete differentiation to CD4+CD8+ preselection stage of development. Erk activity is essential for the process of positive selection, and it differentially affects CD4 and CD8 T cell maturation; yet, diminished expression itself is not sufficient to alter lineage commitment.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Linfócitos T/imunologia , Animais , Relação CD4-CD8 , Proliferação de Células , Deleção de Genes , Inativação Gênica , Subpopulações de Linfócitos/imunologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Mutação , Transdução de Sinais/imunologia , Linfócitos T/citologia , Timo/citologia , Timo/imunologia
8.
Int Immunol ; 17(11): 1379-90, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16172133

RESUMO

DAP kinases are a family of serine/threonine kinases known to regulate intrinsic apoptotic processes. DAP-related apoptotic kinase-2 (DRAK2) is highly expressed in lymphoid organs, with differential expression during thymocyte development. Low levels of transcript were observed in CD4/CD8 double-positive (DP) and double-negative populations, whereas single-positive thymocytes possessed elevated levels. Ex vivo stimulation of DP thymocytes with phorbol myristate acetate or antibodies that activate the TCR complex led to the accumulation of DRAK2 in a protein kinase C- and MAP Kinase-dependent fashion. Although DAP kinase family members are thought to potentiate apoptosis, ectopic expression of DRAK2 using retroviral transduction of primary T cells and NIH3T3 fibroblasts failed to decrease rates of survival, suggesting that DRAK2 expression is not sufficient to promote apoptosis. Rather, our results demonstrate that DRAK2 is a primary response gene activated by TCR stimulation in DP thymocytes. Further, we observed that DRAK2 controlled the threshold for calcium signaling in the thymus since positively selected Drak2-deficient thymocytes displayed a reduced requirement for TCR cross-linking. These findings are consistent with a role for DRAK2 in thymocyte selection and lymphoid maturation, and demonstrate that DRAK2 transduces non-apoptotic signals during thymocyte differentiation.


Assuntos
Apoptose/imunologia , Regulação da Expressão Gênica/imunologia , Ativação Linfocitária/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Sinalização do Cálcio/genética , Sinalização do Cálcio/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Regulação da Expressão Gênica/genética , Ativação Linfocitária/genética , Camundongos , Células NIH 3T3 , Proteínas Serina-Treonina Quinases/genética , Linfócitos T/citologia , Timo/citologia , Timo/imunologia
9.
J Immunol ; 173(5): 3250-60, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15322187

RESUMO

Protein kinase C lambda (PKClambda) is an atypical member of the PKC family of serine/threonine kinases with high similarity to the other atypical family member, PKCzeta. This similarity has made it difficult to determine specific roles for the individual atypical isoforms. Both PKClambda and PKCzeta have been implicated in the signal transduction, initiated by mediators of innate immunity, that culminates in the activation of MAPKs and NF-kappaB. In addition, work from invertebrates shows that atypical PKC molecules play a role in embryo development and cell polarity. To determine the unique functions of PKClambda, mice deficient for PKClambda were generated by gene targeting. The ablation of PKClambda results in abnormalities early in gestation with lethality occurring by embryonic day 9. The role of PKClambda in cytokine-mediated cellular activation was studied by making mouse chimeras from PKClambda-deficient embryonic stem cells and C57BL/6 or Rag2-deficient blastocysts. Cell lines derived from these chimeric animals were then used to dissect the role of PKClambda in cytokine responses. Although the mutant cells exhibited alterations in actin stress fibers and focal adhesions, no other phenotypic differences were noted. Contrary to experiments using dominant interfering forms of PKClambda, mutant cells responded normally to TNF, serum, epidermal growth factor, IL-1, and LPS. In addition, no abnormalities were found in T cell development or T cell activation. These data establish that, in vertebrates, the two disparate functions of atypical PKC molecules have been segregated such that PKCzeta mediates signal transduction of the innate immune system and PKClambda is essential for early embryogenesis.


Assuntos
Deleção de Genes , Isoenzimas/genética , Proteína Quinase C/genética , Sequência de Aminoácidos , Animais , Marcação de Genes , Isoenzimas/fisiologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , NF-kappa B/metabolismo , Proteína Quinase C/deficiência , Proteína Quinase C/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células-Tronco , Linfócitos T/enzimologia , Linfócitos T/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
10.
Int Immunol ; 14(9): 1039-53, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12202401

RESUMO

As a result of a transgene insertion and chromosomal deletion, a mutant mouse strain has been found that is defective in the lineage commitment step that produces a balance of alphabeta and gammadelta T cells. The mice produce a reduced population of alphabeta CD4 T cells and almost no alphabeta CD8 T cells. Within the CD4 and CD8 populations in the thymus there exist abnormal subsets that express the gammadelta TCR. These gammadelta TCR-expressing cells populate the peripheral lymphoid organs such that up to 75% of the CD8 T cells in the lymph nodes and spleen express a gammadelta TCR. Further analyses indicate that the regulation that prevents dual TCR expression has been impaired. The locus of insertion and deletion was mapped to chromosome 10 26 cM. We have analyzed the entire locus and, in addition, the gene expression changes in early thymocytes were analyzed by gene array technology. The analysis of this mutant strain indicates that the alphabeta versus gammadelta lineage decision can be profoundly disregulated independently of successful gene rearrangements.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Animais , Proteínas de Transporte/genética , Integrases/genética , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Timo/anormalidades , Timo/crescimento & desenvolvimento , Proteínas Virais/genética
11.
Mol Cell Biol ; 22(14): 5173-81, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12077344

RESUMO

Macrophage receptors function in pattern recognition for the induction of innate immunity, in cellular communication to mediate the regulation of adaptive immune responses, and in the clearance of some glycosylated cells or glycoproteins from the circulation. They also function in homeostasis by initiating the engulfment of apoptotic cells. Evidence has suggested that macrophage receptors function to recognize cells that are destined for programmed cell death but not yet overtly apoptotic. We have examined the function of a macrophage receptor specific for unsialylated glycoproteins, known as the mouse macrophage galactose- and N-acetylgalactosamine-specific lectin (mMGL) (Ii et al., J. Biol. Chem. 265:11295-11298, 1990; Sato et al., J. Biochem. [Tokyo] 111:331-336, 1992; Yamamoto et al., Biochemistry 33:8159-8166, 1994). With targeted disruption, we tested whether mMGL is necessary for macrophage function, controlled thymic development, the loss of activated CD8 T cells, and the turnover of red blood cells. Evidence indicates that mMGL may play a nonessential role in several of these macrophage functions. Experiments are presented that indicate the existence of another galactose- and N-acetylgalactosamine-recognizing lectin distinct from mMGL. This may explain the absence of a strong phenotype in mMGL-deficient mice.


Assuntos
Hematopoese/fisiologia , Lectinas Tipo C , Lectinas/deficiência , Proteínas de Membrana , Animais , Assialoglicoproteínas , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Envelhecimento Eritrocítico , Contagem de Eritrócitos , Eritropoese/genética , Eritropoese/fisiologia , Feminino , Marcação de Genes , Teste de Complementação Genética , Hematopoese/genética , Homeostase , Lectinas/genética , Lectinas/fisiologia , Tecido Linfoide/crescimento & desenvolvimento , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Sialiltransferases/deficiência , Sialiltransferases/genética , Sialiltransferases/fisiologia , Distribuição Tecidual , beta-Galactosídeo alfa-2,3-Sialiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...