Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Immunology ; 159(2): 205-220, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31642515

RESUMO

Regulatory T (Treg) cells induce immunologic tolerance by suppressing effector functions of conventional lymphocytes in the periphery. On the other hand, immune silencing is mediated by recognition of phosphatidylserine (PS) on apoptotic cells by phagocytes. Here we describe expression of the PS-binding protein Annexin V (ANXA5) in CD4+  CD25hi Treg cells at the mRNA and protein levels. CD4+  ANXA5+ T cells constitute about 0·1%-0·6% of peripheral blood CD3+ T cells, exhibit co-expression of several Treg markers, such as Forkhead box P3, programmed cell death protein-1, cytotoxic T-lymphocyte antigen-4 and CD38. In vitro, ANXA5+ Treg cells showed enhanced adhesion to PS+ endothelial cells. Stimulated by anti-CD3 and PS+ syngeneic antigen-presenting cells CD4+  ANXA5+ T cells expanded in the absence of exogenous interleukin-2. CD4+  ANXA5+ T cells suppressed CD4+  ANXA5- T-cell proliferation and mammalian target of rapamycin phosphorylation, partially dependent on cell contact. CD4+  ANXA5+ T-cell-mediated suppression was allo-specific and accompanied by an increased production of anti-inflammatory mediators. In vivo, using a model of delayed type hypersensitivity, murine CD4+  ANXA5+ T cells inhibited T helper type 1 responses. In conclusion, we report for the first time expression of ANXA5 on a subset of Treg cells that might bridge classical regulatory Treg function with immune silencing.


Assuntos
Anexina A5/metabolismo , Hipersensibilidade Tardia/imunologia , Ativação Linfocitária , Linfócitos T Reguladores/metabolismo , Animais , Anexina A5/genética , Anexina A5/imunologia , Adesão Celular , Proliferação de Células , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Hipersensibilidade Tardia/genética , Hipersensibilidade Tardia/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fenótipo , Fosfatidilserinas/metabolismo , Fosforilação , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Serina-Treonina Quinases TOR/metabolismo , Células Th1/imunologia , Células Th1/metabolismo
2.
Immunity ; 31(4): 665-76, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19818652

RESUMO

The characteristics, importance, and molecular requirements for interactions between mast cells (MCs) and CD8(+) T cells have not been elucidated. Here, we demonstrated that MCs induced antigen-specific CD8(+) T cell activation and proliferation. This process required direct cell contact and MHC class I-dependent antigen cross-presentation by MCs and induced the secretion of interleukin-2, interferon-gamma, and macrophage inflammatory protein-1alpha by CD8(+) T cells. MCs regulated antigen-specific CD8(+) T cell cytotoxicity by increasing granzyme B expression and by promoting CD8(+) T cell degranulation. Because MCs also upregulated their expression of costimulatory molecules (4-1BB) and released osteopontin upon direct T cell contact, MC-T cell interactions probably are bidirectional. In vivo, adoptive transfer of antigen-pulsed MCs induced MHC class I-dependent, antigen-specific CD8(+) T cell proliferation, and MCs regulated CD8(+) T cell-specific priming in experimental autoimmune encephalomyelitis. Thus, MCs are important players in antigen-specific regulation of CD8(+) T cells.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada/imunologia , Encefalomielite Autoimune Experimental/imunologia , Mastócitos/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/microbiologia , Degranulação Celular/imunologia , Quimiocina CCL3/biossíntese , Quimiocina CCL3/imunologia , Técnicas de Cocultura , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Glicoproteínas/imunologia , Granzimas/imunologia , Granzimas/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-2/biossíntese , Interleucina-2/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/microbiologia , Mastócitos/metabolismo , Mastócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Glicoproteína Mielina-Oligodendrócito , Osteopontina/imunologia , Osteopontina/metabolismo , Ovalbumina/imunologia , Fragmentos de Peptídeos/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
3.
Immunol Cell Biol ; 94(1): 109-13, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26150319

RESUMO

The transcription factor hypoxia inducible factor-1α (HIF-1α) mediates the metabolic adaptation of cells to hypoxia and T-helper cell fate. However, HIF-1α regulation in CD4(+) T cells (T cells) remains elusive. Here we observed that depletion of oxygen (O2⩽2%) alone was not sufficient to induce HIF-1α expression in T cells. However, when hypoxic T cells were stimulated, HIF-1α was expressed and this was dependent on nuclear factor-κB- and nuclear factor of activated T cell (NFAT)-mediated transcriptional upregulation of Hif-1α mRNA. HIF-1α upregulation could be blocked by drugs inhibiting NF-κB, NFAT or mammalian target of rapamycin precluding CD4(+) T-cell stimulation or translation in T cells, as well as by blocking transcription. CD3, CD28, phorbol-12-myristat-13-acetat (PMA) or ionomycin-stimulated T cells did not express HIF-1α under normoxic conditions. In conclusion, regulation of HIF-1α expression in CD4(+) T cells in hypoxia gravely relies on its transcriptional upregulation and subsequent enhanced protein stabilization.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Transcrição Gênica , Linfócitos T CD4-Positivos/imunologia , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Ativação Linfocitária/imunologia , Masculino , Estabilidade Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Blood ; 116(15): 2665-75, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20595514

RESUMO

Mast cell (MC) differentiation, survival, and activation are controlled by the membrane tyrosine kinase c-Kit upon interaction with stem cell factor (SCF). Here we describe a single point mutation induced by N-ethyl-N-nitrosurea (ENU) mutagenesis in C57BL/6J mice-an A to T transversion at position 2388 (exon 17) of the c-Kit gene, resulting in the isoleucine 787 substitution by phenylalanine (787F), and analyze the consequences of this mutation for ligand binding, signaling, and MC development. The Kit(787F/787F) mice carrying the single amino acid exchange of c-Kit lacks both mucosal and connective tissue-type MCs. In bone marrow-derived mast cells (BMMCs), the 787F mutation does not affect SCF binding and c-Kit receptor shedding, but strongly impairs SCF-induced cytokine production, degranulation enhancement, and apoptosis rescue. Interestingly, c-Kit downstream signaling in 787F BMMCs is normally initiated (Erk1/2 and p38 activation as well as c-Kit autophosphorylation) but fails to be sustained thereafter. In addition, 787F c-Kit does not efficiently mediate Cbl activation, leading to the absence of subsequent receptor ubiquitination and impaired c-Kit internalization. Thus, I787 provides nonredundant signals for c-Kit internalization and functionality.


Assuntos
Diferenciação Celular/fisiologia , Mastócitos/citologia , Mastócitos/metabolismo , Proteínas Proto-Oncogênicas c-kit/química , Proteínas Proto-Oncogênicas c-kit/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Primers do DNA/genética , Técnicas In Vitro , Interleucina-3/farmacologia , Isoleucina/química , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação Puntual , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais , Fator de Células-Tronco/metabolismo
5.
Immunology ; 131(4): 488-500, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20646075

RESUMO

Symptoms of diseases such as rheumatoid arthritis, which is T helper 1 (Th1) dependent, and asthma, which is T helper 2 (Th2) dependent, are influenced by diurnal rhythms and natural regulatory T cells (nTreg). However, the mechanisms responsible for the diurnal rhythm of disease activity have not been identified and it is unclear whether nTreg activity is diurnal rhythm-dependent. We therefore investigated whether a 24-hr diurnal cycle affected the ability of various helper T-cell populations to generate immunomodulatory and pro-inflammatory cytokines, as well as its suppression by nTreg cells. Using a within-subject crossover design, sleep versus continuous wakefulness was compared over a 24-hr period in healthy young volunteers under defined environmental conditions. Venous blood was drawn periodically every 5 hr and the function of T cells was explored in vitro. We demonstrated that interleukin (IL)-2, interferon-γ (IFN-γ), tumour necrosis factor-α (TNF-α) and IL-10 secretion by naïve CD4(+) T cells follows a diurnal rhythm. Furthermore, multiple regression analysis, as well as subsequent in vitro experiments, suggested that serum levels of cortisol and prolactin are part of the underlying mechanism. Additionally, we observed that nTreg suppressed the secretion of IFN-γ, IL-2 and TNF-α, but not the secretion of IL-4, IL-6, IL-10 and IL-17A. However, the abrogation of IL-2 release was reversed upon inhibiting CD25 on nTreg. Highly purified nTreg secreted IL-6, IL-10 and IL-17A, but not IL-2, IL-4, IFN-γ or TNF-α. Taken together, our results demonstrate that hormones and nTreg modulate the diurnal rhythm of T helper cell activity.


Assuntos
Ritmo Circadiano/imunologia , Citocinas/imunologia , Hormônios/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Asma/imunologia , Asma/metabolismo , Estudos Cross-Over , Citocinas/metabolismo , Hormônios/metabolismo , Humanos , Masculino , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
6.
Gerontology ; 56(6): 574-80, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20130392

RESUMO

The lack of sufficient amounts of sleep is a hallmark of modern living, and it is commonly perceived that in the long run this makes us sick. An increasing amount of scientific data indicate that sleep deprivation has detrimental effects on immune function. Conversely, immune responses feedback on sleep phase and architecture. Several studies have investigated the impact of short-term sleep deprivation on different immune parameters, whereas only a few studies have addressed the influence of sleep restriction on the immune system. In many cases, sleep deprivation and restriction impair immune responses by disrupting circadian rhythms at the level of immune cells, which might be a consequence of disrupted endocrine and physiological circadian rhythms. Little is known about the mechanisms underlying the circadian regulation of immunity, but recent studies have suggested that local as well as central circadian clocks drive the rhythms of immune function. In this review, we present a mechanistic model which proposes that sleep (through soluble factors and body temperature) primes immune cells on the one hand, and, on the other hand, provides a timing signal for hematopoietic circadian clocks. We hypothesize that chronic sleep disruption desynchronizes these clocks and, through this mechanism, deregulates immune responses.


Assuntos
Sistema Imunitário/patologia , Imunidade , Modelos Biológicos , Privação do Sono/imunologia , Sono , Animais , Temperatura Corporal/imunologia , Ritmo Circadiano/imunologia , Retroalimentação Fisiológica , Feminino , Hematopoese/imunologia , Hormônios/imunologia , Humanos , Sistema Imunitário/fisiologia , Estilo de Vida , Masculino , Neuroimunomodulação , Sono/imunologia , Privação do Sono/psicologia
7.
J Leukoc Biol ; 96(2): 305-12, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24664971

RESUMO

The central oxygen sensitive transcription factor HIF-1α has been implicated in the differentiation of n(T(reg)) and Th17 cells and to orchestrate metabolic changes of activated T cells. However, data on the functional relevance of HIF-1α and Hox, in general, for nT(reg)-suppressive activity and T cell function in primary human cells are still missing. Therefore, we analyzed the effect of Hox and HIF-1α on human T(res), n(Treg), and Th17 cells. Under Hox, nT(reg)-mediated suppression of T(res) proliferation, CD25 expression, and secretion of IFN-γ were significantly reduced, whereas expression levels of VEGF, TNF-α, and IL-10 were significantly increased. In contrast to observations in mice, Th17 lineage commitment, as determined by RORγt expression, was not affected by activation or inhibition of HIF-1α expression using DMOG or YC-1 treatment, respectively. Nevertheless, the secretion of IL-17A was increased by DMOG and reduced by YC-1 under Th17-skewing conditions in a dose- dependent manner. In conclusion, Hox and HIF-1α substantially influence human T cell-mediated immune responses by modulation of nT(reg)-suppressive function and IL-17A secretion by Th17 cells.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Antígenos CD4/imunologia , Citocinas/imunologia , Ativadores de Enzimas/farmacologia , Feminino , Humanos , Indazóis/farmacologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Masculino , Camundongos , Linfócitos T Reguladores/citologia , Células Th17/citologia
8.
PLoS One ; 7(9): e45234, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028866

RESUMO

Regulatory CD8(+) T cells are critical for self-tolerance and restricting excessive immune responses. The variety of immune functions they fulfill, the heterogeneity of their phenotype, and the mechanism of action are still poorly understood. Here we describe that regulatory CD8(+) T cells exhibiting immunosuppressive actions in vitro and in vivo are recognized as CD38(high) T cells and present in naive mice. CD38 is a glycosylated membrane protein with ectonucleotidase properties. CD8(+)CD38(high) (CD44(+)CD122(+)CD62L(high)) lymphocytes suppress CD4(+) effector T-cell proliferation in an antigen-non specific manner via IFN-γ. While direct cell-to-cell contact is needed for this suppressor activity, it is independent of membrane-bound TGF-ß and granzyme B release. IL-15 potentiates the suppressive activity of CD8(+)CD38(high) T cells and controls their survival and expansion. In humans CD8(+)CD38(high) T cells inhibit CD4(+) effector T cell proliferation. In vivo, CD8(+)CD38(high), but not CD8(+)CD38(-) T cells mitigate murine experimental autoimmune encephalomyelitis (EAE) by reducing the clinical score and delaying disease occurrence. EAE suppression is enhanced by pre-treatment of CD8(+)CD38(high) T cells with IL-15. These findings add evidence that the expression of ectoenzyme receptor family members positively correlates with suppressor functions and identifies CD8(+)CD38(high) T cells as potential inhibitors of excessive immune responses.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Linfócitos T CD8-Positivos/patologia , Encefalomielite Autoimune Experimental/patologia , Interferon gama/imunologia , Glicoproteínas de Membrana/imunologia , Nucleotidases/imunologia , Linfócitos T Reguladores/patologia , ADP-Ribosil Ciclase 1/metabolismo , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Comunicação Celular/imunologia , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Citometria de Fluxo , Memória Imunológica , Imunofenotipagem , Interleucina-15/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nucleotidases/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
9.
Proc Natl Acad Sci U S A ; 103(37): 13837-42, 2006 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-16945916

RESUMO

The obligate intracellular pathogen Leishmania major survives and multiplies in professional phagocytes. The evasion strategy to circumvent killing by host phagocytes and establish a productive infection is poorly understood. Here we report that the virulent inoculum of Leishmania promastigotes contains a high ratio of annexin A5-binding apoptotic parasites. This subpopulation of parasites is characterized by a round body shape, a swollen kinetoplast, nuclear condensation, and a lack of multiplication and represents dying or already dead parasites. After depleting the apoptotic parasites from a virulent population, Leishmania do not survive in phagocytes in vitro and lose their disease-inducing ability in vivo. TGF-beta induced by apoptotic parasites is likely to mediate the silencing of phagocytes and lead to survival of infectious Leishmania populations. The data demonstrate that apoptotic promastigotes, in an altruistic way, enable the intracellular survival of the viable parasites.


Assuntos
Anexina A5/metabolismo , Leishmania major/citologia , Leishmania major/patogenicidade , Leishmaniose/imunologia , Fagócitos/parasitologia , Animais , Anexina A5/análise , Apoptose , Regulação para Baixo , Feminino , Leishmania major/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fator de Crescimento Transformador alfa/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , Virulência
10.
J Immunol ; 172(3): 1768-76, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14734760

RESUMO

The obligate intracellular bacterial pathogen Chlamydia pneumoniae (Cp) is responsible for a range of human diseases, including acute respiratory infection. Although experimental intratracheal infection with Cp results in a massive recruitment of neutrophil granulocytes (polymorphonuclear neutrophils (PMN)), the role of these cells in the defense against Cp is unclear. In this study the interactions of PMN with Cp were investigated. In vitro coincubation experiments showed that human granulocytes were able to internalize Chlamydia in an opsonin-independent manner. Importantly, phagocytosed Cp were not killed; the ingested bacteria survived and multiplied within PMN. Although uninfected granulocytes became apoptotic within 10 h, infected PMN survived up to 90 h. Coincubation with Cp significantly decreased the ratio of apoptotic PMN, as detected by morphological analysis, annexin V, and TUNEL staining. The observed antiapoptotic effect was associated with a markedly lower level of procaspase-3 processing and, consequently, reduced caspase-3 activity in infected PMN. LPS was found as a major, but not exclusive, component responsible for the observed antiapoptotic effect. Chlamydia LPS affected PMN apoptosis both by acting directly on the cells and by inducing the autocrine production of the antiapoptotic cytokine IL-8. These data show that, in contrast to other microbial pathogens that drive phagocytes into apoptosis to escape killing, Cp can extend the life span of neutrophil granulocytes, making them suitable host cells for survival and multiplication within the first hours/days after infection.


Assuntos
Apoptose/imunologia , Chlamydophila pneumoniae/crescimento & desenvolvimento , Neutrófilos/microbiologia , Adulto , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Comunicação Celular/imunologia , Sobrevivência Celular/imunologia , Sistema Livre de Células/imunologia , Sistema Livre de Células/microbiologia , Células Cultivadas , Chlamydophila pneumoniae/patogenicidade , Técnicas de Cocultura , Regulação para Baixo/imunologia , Precursores Enzimáticos/antagonistas & inibidores , Precursores Enzimáticos/metabolismo , Temperatura Alta , Humanos , Interleucina-8/metabolismo , Interleucina-8/farmacologia , Líquido Intracelular/imunologia , Líquido Intracelular/microbiologia , Lipopolissacarídeos/farmacologia , Neutrófilos/citologia , Neutrófilos/enzimologia , Neutrófilos/metabolismo , Fagocitose/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Proteínas Recombinantes/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA