Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(13): e2306763121, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38498711

RESUMO

Lactate-proton symporter monocarboxylate transporter 1 (MCT1) facilitates lactic acid export from T cells. Here, we report that MCT1 is mandatory for the development of virus-specific CD8+ T cell memory. MCT1-deficient T cells were exposed to acute pneumovirus (pneumonia virus of mice, PVM) or persistent γ-herpesvirus (Murid herpesvirus 4, MuHV-4) infection. MCT1 was required for the expansion of virus-specific CD8+ T cells and the control of virus replication in the acute phase of infection. This situation prevented the subsequent development of virus-specific T cell memory, a necessary step in containing virus reactivation during γ-herpesvirus latency. Instead, persistent active infection drove virus-specific CD8+ T cells toward functional exhaustion, a phenotype typically seen in chronic viral infections. Mechanistically, MCT1 deficiency sequentially impaired lactic acid efflux from activated CD8+ T cells, caused an intracellular acidification inhibiting glycolysis, disrupted nucleotide synthesis in the upstream pentose phosphate pathway, and halted cell proliferation which, ultimately, promoted functional CD8+ T cell exhaustion instead of memory development. Taken together, our data demonstrate that MCT1 expression is mandatory for inducing T cell memory and controlling viral infection by CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos , Transportadores de Ácidos Monocarboxílicos , Simportadores , Animais , Camundongos , Transporte Biológico , Linfócitos T CD8-Positivos/metabolismo , Ácido Láctico/metabolismo , Simportadores/genética , Simportadores/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo
2.
Cell Rep ; 42(8): 112876, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37543948

RESUMO

The co-inhibitory programmed death (PD)-1 signaling pathway plays a major role in the context of tumor-specific T cell responses. Conversely, it also contributes to the maintenance of peripheral tolerance, as patients receiving anti-PD-1 treatment are prone to developing immune-related adverse events. Yet, the physiological role of the PD-1/PDL-1 axis in T cell homeostasis is still poorly understood. Herein, we show that under steady-state conditions, the absence of PD-1 signaling led to a preferential expansion of CD8+ T cells in the liver. These cells exhibit an oligoclonal T cell receptor (TCR) repertoire and a terminally differentiated exhaustion profile. The transcription factor EOMES is required for the clonal expansion and acquisition of this differentiation program. Finally, single-cell transcriptomics coupled with TCR repertoire analysis support the notion that these cells arise locally from liver-resident memory CD8+ T cells. Overall, we show a role for PD-1 signaling in liver memory T cell homeostasis.


Assuntos
Linfócitos T CD8-Positivos , Regulação da Expressão Gênica , Humanos , Linfócitos T CD8-Positivos/metabolismo , Fígado/metabolismo , Transdução de Sinais , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
3.
EMBO Rep ; 24(7): e56460, 2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37144276

RESUMO

Hypoxia induces profound modifications in the gene expression program of eukaryotic cells due to lowered ATP supply resulting from the blockade of oxidative phosphorylation. One significant consequence of oxygen deprivation is the massive repression of protein synthesis, leaving a limited set of mRNAs to be translated. Drosophila melanogaster is strongly resistant to oxygen fluctuations; however, the mechanisms allowing specific mRNA to be translated into hypoxia are still unknown. Here, we show that Ldh mRNA encoding lactate dehydrogenase is highly translated into hypoxia by a mechanism involving a CA-rich motif present in its 3' untranslated region. Furthermore, we identified the cap-binding protein eIF4EHP as a main factor involved in 3'UTR-dependent translation under hypoxia. In accordance with this observation, we show that eIF4EHP is necessary for Drosophila development under low oxygen concentrations and contributes to Drosophila mobility after hypoxic challenge. Altogether, our data bring new insight into mechanisms contributing to LDH production and Drosophila adaptation to oxygen variations.


Assuntos
Drosophila melanogaster , Hipóxia , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Hipóxia/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Drosophila/genética , Drosophila/metabolismo , Oxigênio/metabolismo , Regiões 3' não Traduzidas , L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/metabolismo , Biossíntese de Proteínas
4.
Elife ; 112022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35192456

RESUMO

The oxygen sensor prolyl hydroxylase domain 2 (PHD2) plays an important role in cell hypoxia adaptation by regulating the stability of HIF proteins (HIF1α and HIF2α) in numerous cell types, including T lymphocytes. The role of oxygen sensor on immune cells, particularly on regulatory T cell (Treg) function, has not been fully elucidated. The purpose of our study was to evaluate the role of PHD2 in the regulation of Treg phenotype and function. We demonstrate herein that selective ablation of PHD2 expression in Treg (PHD2ΔTreg mice) leads to a spontaneous systemic inflammatory syndrome, as evidenced by weight loss, development of a rectal prolapse, splenomegaly, shortening of the colon, and elevated expression of IFN-γ in the mesenteric lymph nodes, intestine, and spleen. PHD2 deficiency in Tregs led to an increased number of activated CD4 conventional T cells expressing a Th1-like effector phenotype. Concomitantly, the expression of innate-type cytokines such as Il1b, Il12a, Il12b, and Tnfa was found to be elevated in peripheral (gut) tissues and spleen. PHD2ΔTreg mice also displayed an enhanced sensitivity to dextran sodium sulfate-induced colitis and toxoplasmosis, suggesting that PHD2-deficient Tregs did not efficiently control inflammatory response in vivo, particularly those characterized by IFN-γ production. Further analysis revealed that Treg dysregulation was largely prevented in PHD2-HIF2α (PHD2-HIF2αΔTreg mice), but not in PHD2-HIF1α (PHD2-HIF1αΔTreg mice) double KOs, suggesting an important and possibly selective role of the PHD2-HIF2α axis in the control of Treg function. Finally, the transcriptomic analysis of PHD2-deficient Tregs identified the STAT1 pathway as a target of the PHD2-HIF2α axis in regulatory T cell phenotype and in vivo function.


Assuntos
Colite , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Linfócitos T Reguladores , Animais , Colite/induzido quimicamente , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Oxigênio , Pró-Colágeno-Prolina Dioxigenase , Prolil Hidroxilases
5.
Sci Rep ; 10(1): 21673, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33303820

RESUMO

The AMP-activated kinase (AMPK) is a major energy sensor metabolic enzyme that is activated early during T cell immune responses but its role in the generation of effector T cells is still controversial. Using both in vitro and in vivo models of T cell proliferation, we show herein that AMPK is dispensable for early TCR signaling and short-term proliferation but required for sustained long-term T cell proliferation and effector/memory T cell survival. In particular, AMPK promoted accumulation of effector/memory T cells in competitive homeostatic proliferation settings. Transplantation of AMPK-deficient hematopoïetic cells into allogeneic host recipients led to a reduced graft-versus-host disease, further bolstering a role for AMPK in the expansion and pathogenicity of effector T cells. Mechanistically, AMPK expression enhances the mitochondrial membrane potential of T cells, limits reactive oxygen species (ROS) production, and resolves ROS-mediated toxicity. Moreover, dampening ROS production alleviates the proliferative defect of AMPK-deficient T cells, therefore indicating a role for an AMPK-mediated ROS control of T cell fitness.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proliferação de Células/genética , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/imunologia , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/fisiologia , Sobrevivência Celular/genética , Células Cultivadas , Expressão Gênica , Humanos , Potencial da Membrana Mitocondrial , Espécies Reativas de Oxigênio/toxicidade , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais
6.
Front Immunol ; 11: 206, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117317

RESUMO

Beyond its well-admitted role in development and organogenesis, it is now clear that the transcription factor c-Maf has owned its place in the realm of immune-related transcription factors. Formerly introduced solely as a Th2 transcription factor, the role attributed to c-Maf has gradually broadened over the years and has extended to most, if not all, known immune cell types. The influence of c-Maf is particularly prominent among T cell subsets, where c-Maf regulates the differentiation as well as the function of multiple subsets of CD4 and CD8 T cells, lending it a crucial position in adaptive immunity and anti-tumoral responsiveness. Recent research has also revealed the role of c-Maf in controlling Th17 responses in the intestine, positioning it as an essential factor in intestinal homeostasis. This review aims to present and discuss the recent advances highlighting the particular role played by c-Maf in T lymphocyte differentiation, function, and homeostasis.


Assuntos
Tolerância Imunológica , Proteínas Proto-Oncogênicas c-maf/fisiologia , Linfócitos T/imunologia , Diferenciação Celular , Humanos , Interleucina-10/biossíntese , Intestinos/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/análise , Proteínas Proto-Oncogênicas c-maf/genética , Linfócitos T/citologia , Linfócitos T/fisiologia
7.
Front Immunol ; 10: 3007, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31998303

RESUMO

RORγt-expressing Tregs form a specialized subset of intestinal CD4+ Foxp3+ cells which is essential to maintain gut homeostasis and tolerance to commensal microbiota. Recently, c-Maf emerged as a critical factor in the regulation of RORγt expression in Tregs. However, aside from c-Maf signaling, the signaling pathways involved in the differentiation of RORγt+ Tregs and their possible interplay with c-Maf in this process are largely unknown. We show that RORγt+ Treg development is controled by positive as well as negative signals. Along with c-Maf signaling, signals derived from a complex microbiota, as well as IL-6/STAT3- and TGF-ß-derived signals act in favor of RORγt+ Treg development. Ectopic expression of c-Maf did not rescue RORγt expression in STAT3-deficient Tregs, indicating the presence of additional effectors downstream of STAT3. Moreover, we show that an inflammatory IFN-γ/STAT1 signaling pathway acts as a negative regulator of RORγt+ Treg differentiation in a c-Maf independent fashion. These data thus argue for a complex integrative signaling network that finely tunes RORγt expression in Tregs. The finding that type 1 inflammation impedes RORγt+ Treg development even in the presence of an active IL-6/STAT3 pathway further suggests a dominant negative effect of STAT1 over STAT3 in this process.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/imunologia , Expressão Gênica , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Imunofenotipagem , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-maf/genética , Proteínas Proto-Oncogênicas c-maf/metabolismo , Fator de Transcrição STAT3/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/citologia , Fator de Crescimento Transformador beta/metabolismo
8.
Methods Mol Biol ; 1732: 465-476, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29480493

RESUMO

Distinct lymphocyte subpopulations display discrete metabolic profiles and are differently affected by metabolic resource variations, making the analysis of lymphocyte survival in a complex tissue in response to metabolic stress highly challenging. Here we describe a flow cytometry-based method allowing simultaneous cell identification and viable cell counting in mixed lymphocyte populations without extensive cell subset purification procedures. The example provided herein illustrates the role of AMPK in T lymphocyte survival in response to the mitochondrial poison oligomycin.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Citometria de Fluxo/métodos , Estresse Fisiológico/fisiologia , Subpopulações de Linfócitos T/fisiologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Contagem de Células/instrumentação , Contagem de Células/métodos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citometria de Fluxo/instrumentação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Oligomicinas/farmacologia , Baço/citologia , Estresse Fisiológico/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/metabolismo
9.
Front Immunol ; 8: 480, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28496444

RESUMO

Follicular helper T cells (Tfh) have been identified as the primary cell subpopulation regulating B cell responses in germinal centers, thus supporting high-affinity antibody production. Among the transcription factors orchestrating Tfh cell differentiation and function, the role played by the proto-oncogene c-Maf remains poorly characterized. We report herein that selective loss of c-Maf expression in the T cell compartment results in defective development of Tfh cells in response to both antigen/adjuvant vaccinations and commensal intestinal bacteria. Accordingly, c-Maf expression in T cells was essential for the development and high-affinity antibody secretion in vaccinated animals. c-Maf was expressed early, concomitantly to BCL6, in Tfh cell precursors and found to regulate Tfh fate in a cell-autonomous fashion. Altogether, our findings reveal a novel, non-redundant, function for c-Maf in the differentiation of Tfh cells and the regulation of humoral immune responses to T-cell-dependent antigens.

10.
J Leukoc Biol ; 101(1): 5-14, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27474166

RESUMO

Follicular helper T cells (Tfh) support high-affinity Ab production by germinal center B cells through both membrane interactions and secretion of IL-4 and -21, two major cytokines implicated in B-cell survival and Ab class switch. Tfh-2 cells recently emerged in humans as a strong IL-4 producer Tfh cell subset implicated in both autoimmune and allergic diseases. Although the molecular mechanisms governing Tfh cell differentiation from naive T cells have been widely described, much less is known about the regulation of cytokine secretion by mouse Tfh-2 cells. The purpose of our study was to evaluate the role of dendritic cell-derived IL-6 in fine-tuning cytokine secretion by Tfh cells. Our results demonstrate that priming of Th cells by IL-6-deficient antigen-presenting dendritic cells preferentially leads to accumulation of a subset of Tfh cells characterized by high expression of GATA3 and IL-4, associated with reduced production of IL-21. STAT3-deficient Tfh cells also overexpress GATA3, suggesting that early IL-6/STAT3 signaling during Tfh cell development inhibits the expression of a set of genes associated with the Th2 differentiation program. Overall, our data indicate that IL-6/STAT3 signaling restrains the expression of Th2-like genes in Tfh cells, thus contributing to the control of IgE secretion in vivo.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Fator de Transcrição GATA3/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Animais , Antígenos/imunologia , Linfócitos B/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Imunização , Switching de Imunoglobulina , Imunoglobulina E/metabolismo , Interleucina-12/metabolismo , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Haematologica ; 100(8): 1023-30, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25934767

RESUMO

Hyaluronan is a major component of the extracellular matrix and glycocalyx. Its main somatic degrading enzymes are hyaluronidases 1 and 2, neither of which is active in the bloodstream. We generated hyaluronidase 2-deficient mice. These animals suffer from chronic, mild anemia and thrombocytopenia, in parallel with a 10-fold increase in plasma hyaluronan concentration. In this study we explored the mechanism of these hematologic anomalies. The decreased erythrocyte and platelet counts were attributed to peripheral consumption. The erythrocyte half-life was reduced from 25 to 8 days without signs of premature aging. Hyaluronidase 2-deficient platelets were functional. Major intrinsic defects in erythrocyte membrane or stability, as well as detrimental effects of high hyaluronan levels on erythrocytes, were ruled out in vitro. Normal erythrocytes transfused into hyaluronidase 2-deficient mice were quickly destroyed but neither splenectomy nor anti-C5 administration prevented chronic hemolysis. Schistocytes were present in blood smears from hyaluronidase 2-deficient mice at a level of 1% to 6%, while virtually absent in control mice. Hyaluronidase 2-deficient mice had increased markers of endothelial damage and microvascular fibrin deposition, without renal failure, accumulation of ultra-large multimers of von Willebrand factor, deficiency of A Disintegrin And Metalloproteinase with ThromboSpondin type 1 motifs, member 13 (ADAMTS13), or hypertension. There was no sign of structural damage in hepatic or splenic sinusoids, or in any other microvessels. We conclude that hyaluronidase 2 deficiency induces chronic thrombotic microangiopathy with hemolytic anemia in mice. The link between this uncommon condition and hyaluronidase 2 remains to be explored in humans.


Assuntos
Hialuronoglucosaminidase/deficiência , Microangiopatias Trombóticas/genética , Proteína ADAMTS13 , Anemia Macrocítica/sangue , Anemia Macrocítica/genética , Animais , Viscosidade Sanguínea/genética , Transplante de Medula Óssea , Sobrevivência Celular/genética , Senescência Celular/genética , Modelos Animais de Doenças , Índices de Eritrócitos , Transfusão de Eritrócitos , Eritrócitos/citologia , Eritrócitos/metabolismo , Eritrócitos Anormais/metabolismo , Proteínas Ligadas por GPI/deficiência , Ácido Hialurônico/sangue , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Knockout , Trombocitopenia/sangue , Trombocitopenia/genética , Microangiopatias Trombóticas/diagnóstico , Microangiopatias Trombóticas/terapia
12.
Int Rev Immunol ; 34(1): 67-81, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25360847

RESUMO

Adenosine monophosphate-activated protein kinase (AMPK) is a serine/threonine kinase that is crucial for cellular energy metabolism homeostasis. AMPK monitors cellular energy status in response to nutritional variations and, once activated by low energy status, switches on ATP-producing catabolic pathways and switches off ATP-consuming anabolic pathways to restore cellular energy homeostasis. When T lymphocytes encounter foreign antigens, they initiate a program of differentiation leading to the rapid generation of effector and memory cells that clear the pathogen and prevent future infection, respectively. Differentiation of naïve T cells in effector or long term memory cells is tightly associated with changes in their energy metabolic activity and recent data have revealed that fine-tuning of metabolism could modulate T cell functions. Here, we will review recent data about the regulation of T cell metabolism by AMPK and discuss its influence on T cell function.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Metabolismo Energético/imunologia , Subunidades Proteicas/metabolismo , Subpopulações de Linfócitos T/enzimologia , Linfócitos T/enzimologia , Proteínas Quinases Ativadas por AMP/química , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/imunologia , Trifosfato de Adenosina/metabolismo , Diferenciação Celular , Metabolismo Energético/genética , Regulação da Expressão Gênica , Homeostase/imunologia , Humanos , Memória Imunológica , Ativação Linfocitária , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/imunologia , Transdução de Sinais , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia
13.
Eur J Immunol ; 44(11): 3252-62, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25092208

RESUMO

The identification of DC-derived signals orchestrating activation of Th1 and Th17 immune responses has advanced our understanding on how these inflammatory responses develop. However, whether specific signals delivered by DCs also participate in the regulation of Th2 immune responses remains largely unknown. In this study, we show that administration of antigen-loaded, IL-6-deficient DCs to naïve mice induced an exacerbated Th2 response, characterized by the differentiation of GATA-3-expressing T lymphocytes secreting high levels of IL-4, IL-5, and IL-13. Coinjection of wild type and IL-6-deficient bone marrow-derived dendritic cells (BMDCs) confirmed that IL-6 exerted a dominant, negative influence on Th2-cell development. This finding was confirmed in vitro, where exogenously added IL-6 was found to limit IL-4-induced Th2-cell differentiation. iNKT cells were required for optimal Th2-cell differentiation in vivo although their activation occurred independently of IL-6 secretion by the BMDCs. Collectively, these observations identify IL-6 secretion as a major, unsuspected, mechanism whereby DCs control the magnitude of Th2 immunity.


Assuntos
Células Dendríticas/imunologia , Interleucina-6/imunologia , Células Th2/citologia , Animais , Asma/imunologia , Basófilos/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Diferenciação Celular , Células Dendríticas/citologia , Células Dendríticas/transplante , Fator de Transcrição GATA3/biossíntese , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Interleucina-6/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/imunologia , Ovalbumina , Células Th2/imunologia
14.
Eur J Immunol ; 43(6): 1489-98, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23504518

RESUMO

CD4(+) T-cell help for B cells is crucial for effective Ab responses. Although follicular T helper (Tfh) cells have emerged as the main providers of T-cell help to B lymphocytes during the germinal center reaction, much less is known about the helper capacities of other effector CD4(+) T cells. The purpose of the present study was to evaluate the acquisition of B-cell help capacity of canonically derived T helper 2 (Th2) cells, a Th-cell subset originally considered responsible for B-cell help in vivo. We demonstrate herein that developing Th2 cells in mice co-express activated forms of signal transducer and activator of transcription 6 (STAT6) and STAT3 and that STAT3 expression was required for the capacity of Th2 cells to provide B-cell help. Thus, Th2 lymphocytes share a common, STAT3-mediated activation program for the acquisition of optimal B-cell help capacity with Tfh cells. Moreover, the expression of STAT3 in Th2 cells enhanced the IgG1-to-IgE class switch ratio in vivo, a finding with important implications for understanding the molecular basis of allergic diseases.


Assuntos
Linfócitos B/imunologia , Ativação Linfocitária , Fator de Transcrição STAT3/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th2/imunologia , Animais , Comunicação Celular , Células Cultivadas , Switching de Imunoglobulina/genética , Imunoglobulina E/genética , Imunoglobulina G/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/imunologia , Ativação Transcricional/genética
15.
PLoS One ; 8(12): e84415, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386375

RESUMO

Follicular T helper cells (Tfh) are crucial for effective antibody responses and long term T cell-dependent humoral immunity. Although many studies are devoted to this novel T helper cell population, the molecular mechanisms governing Tfh cell differentiation have yet to be characterized. MyoR/ABF-1 is a basic helix-loop-helix transcription factor that plays a role in the differentiation of the skeletal muscle and Hodgkin lymphoma. Here we show that MyoR mRNA is progressively induced during the course of Tfh-like cell differentiation in vitro and is expressed in Tfh responding to Alum-precipitated antigens in vivo. This expression pattern suggests that MyoR could play a role in the differentiation and/or function of Tfh cells. We tested this hypothesis using MyoR-deficient mice and found this deficiency had no impact on Tfh differentiation. Hence, MyoR-deficient mice developed optimal T-dependent humoral responses to Alum-precipitated antigens. In conclusion, MyoR is a transcription factor selectively up-regulated in CD4 T cells during Tfh cell differentiation in vitro and upon response to alum-protein vaccines in vivo, but the functional significance of this up-regulation remains uncertain.


Assuntos
Diferenciação Celular/fisiologia , RNA Mensageiro/biossíntese , Linfócitos T Auxiliares-Indutores/metabolismo , Fatores de Transcrição/biossíntese , Regulação para Cima/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
16.
J Immunol ; 187(9): 4517-29, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21948987

RESUMO

Sirtuins are a unique class of NAD(+)-dependent deacetylases that regulate diverse biological functions such as aging, metabolism, and stress resistance. Recently, it has been shown that sirtuins may have anti-inflammatory activities by inhibiting proinflammatory transcription factors such as NF-κB. In contrast, we report in this study that pharmacological inhibition of sirtuins dampens adaptive Th2 responses and subsequent allergic inflammation by interfering with lung dendritic cell (DC) function in a mouse model of airway allergy. Using genetic engineering, we demonstrate that sirtuin 1 represses the activity of the nuclear receptor peroxisome proliferator-activated receptor-γ in DCs, thereby favoring their maturation toward a pro-Th2 phenotype. This study reveals a previously unappreciated function of sirtuin 1 in the regulation of DC function and Th2 responses, thus shedding new light on our current knowledge on the regulation of inflammatory processes by sirtuins.


Assuntos
Asma/imunologia , Células Dendríticas/imunologia , PPAR gama/antagonistas & inibidores , Sirtuína 1/fisiologia , Células Th2/imunologia , Animais , Asma/enzimologia , Asma/patologia , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Feminino , Imunofenotipagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , PPAR gama/metabolismo , Sirtuína 1/antagonistas & inibidores , Células Th2/enzimologia , Células Th2/patologia
17.
J Immunol ; 186(4): 2245-53, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21248260

RESUMO

Adjuvant formulations boost humoral responses by acting through several, yet incompletely elucidated pathways. In this study, we show that oligomycin or 5-aminoimidazole-4-carboxamide-1-ß-D-ribonucleoside (AICAR) enhances Ab production when coinjected with T cell-dependent Ags. Oligomycin and AICAR lead to intracellular ATP reduction, suggesting that metabolic stress could be sensed by immune cells and leads to increased humoral responses. AICAR promotes IL-4 and IL-21 by naive Th cells but does not affect dendritic cell activation/maturation in vitro or in vivo. Accordingly, the adjuvant effect of AICAR or oligomycin does not require MyD88 or caspase-1 expression in vivo. Because AICAR is well tolerated in humans, this compound could represent a novel and safe adjuvant promoting humoral responses in vivo with a minimal reactogenicity.


Assuntos
Imunoglobulina G/biossíntese , Inflamassomos/metabolismo , Estresse Fisiológico/imunologia , Trifosfato de Adenosina/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Adjuvantes Imunológicos/farmacologia , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Inflamassomos/fisiologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Oligomicinas/farmacologia , Ribonucleotídeos/farmacologia , Regulação para Cima/imunologia
18.
FEBS Lett ; 584(16): 3667-71, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20670625

RESUMO

AMP-activated protein kinase (AMPK) plays a pivotal role in regulating cellular energy metabolism. We previously showed that AMPKalpha1-/- mice develop moderate anemia associated with splenomegaly and high reticulocytosis. Here, we report that splenectomy of AMPKalpha1-/- mice worsened anemia supporting evidence that AMPKalpha1-/- mice developed a compensatory response through extramedullary erythropoiesis in the spleen. Transplantation of bone marrow from AMPKalpha1-/- mice into wild-type recipients recapitulated the hematologic phenotype. Further, AMPKalpha1-/- red blood cells (RBC) showed less deformability in response to shear stress limiting their membrane flexibility. Thus, our results highlight the crucial role of AMPK to preserve RBC integrity.


Assuntos
Proteínas Quinases Ativadas por AMP/sangue , Deformação Eritrocítica/fisiologia , Proteínas Quinases Ativadas por AMP/deficiência , Proteínas Quinases Ativadas por AMP/genética , Anemia/sangue , Anemia/enzimologia , Anemia/genética , Animais , Transplante de Medula Óssea , Deformação Eritrocítica/genética , Eritropoese , Hematopoese Extramedular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragilidade Osmótica , Esplenectomia , Esplenomegalia/sangue , Esplenomegalia/enzimologia , Esplenomegalia/genética
19.
Cancer Res ; 70(1): 8-11, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20028851

RESUMO

Beyond its well-described role in cellular metabolism, intracellular nicotinamide adenine dinucleotide (NAD) levels have been shown to affect the enzymatic activity of a series of NAD-dependent enzymes, influencing biological responses such as cell survival and inflammation. Nicotinamide phosphoribosyl transferase activity has been shown to be essential for maintaining adequate intracellular NAD levels, suggesting that this enzyme may in fact play a central role in modulating the activity of a wide range of NAD-dependent enzymes. Several recent observations concur with this hypothesis and suggest that by regulating NAD availability, Nampt is able to control both cell viability and the inflammatory response. Nampt may thus represent a novel pharmacological target with valuable anti-inflammatory and antitumor properties.


Assuntos
Inflamação/metabolismo , Neoplasias/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Animais , Humanos , NAD/metabolismo
20.
PLoS Pathog ; 5(12): e1000685, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19997494

RESUMO

Apolipoprotein L-I (apoL1) is a human-specific serum protein that kills Trypanosoma brucei through ionic pore formation in endosomal membranes of the parasite. The T. brucei subspecies rhodesiense and gambiense resist this lytic activity and can infect humans, causing sleeping sickness. In the case of T. b. rhodesiense, resistance to lysis involves interaction of the Serum Resistance-Associated (SRA) protein with the C-terminal helix of apoL1. We undertook a mutational and deletional analysis of the C-terminal helix of apoL1 to investigate the linkage between interaction with SRA and lytic potential for different T. brucei subspecies. We confirm that the C-terminal helix is the SRA-interacting domain. Although in E. coli this domain was dispensable for ionic pore-forming activity, its interaction with SRA resulted in inhibition of this activity. Different mutations affecting the C-terminal helix reduced the interaction of apoL1 with SRA. However, mutants in the L370-L392 leucine zipper also lost in vitro trypanolytic activity. Truncating and/or mutating the C-terminal sequence of human apoL1 like that of apoL1-like sequences of Papio anubis resulted in both loss of interaction with SRA and acquired ability to efficiently kill human serum-resistant T. b. rhodesiense parasites, in vitro as well as in transgenic mice. These findings demonstrate that SRA interaction with the C-terminal helix of apoL1 inhibits its pore-forming activity and determines resistance of T. b. rhodesiense to human serum. In addition, they provide a possible explanation for the ability of Papio serum to kill T. b. rhodesiense, and offer a perspective to generate transgenic cattle resistant to both T. b. brucei and T. b. rhodesiense.


Assuntos
Apolipoproteínas/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Lipoproteínas HDL/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Protozoários/metabolismo , Trypanosoma brucei brucei/fisiologia , Trypanosoma brucei rhodesiense/fisiologia , Sequência de Aminoácidos , Animais , Apolipoproteína L1 , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas/farmacologia , Análise Mutacional de DNA , Humanos , Zíper de Leucina/genética , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/farmacologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Papio anubis , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/farmacologia , Ligação Proteica , Alinhamento de Sequência , Termodinâmica , Tripanossomicidas/metabolismo , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei rhodesiense/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA