Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Annu Rev Immunol ; 30: 39-68, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22136167

RESUMEN

mTOR is an evolutionarily conserved serine/threonine kinase that plays a central role in integrating environmental cues in the form of growth factors, amino acids, and energy. In the study of the immune system, mTOR is emerging as a critical regulator of immune function because of its role in sensing and integrating cues from the immune microenvironment. With the greater appreciation of cellular metabolism as an important regulator of immune cell function, mTOR is proving to be a vital link between immune function and metabolism. In this review, we discuss the ability of mTOR to direct the adaptive immune response. Specifically, we focus on the role of mTOR in promoting differentiation, activation, and function in T cells, B cells, and antigen-presenting cells.


Asunto(s)
Inmunidad , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Activación Enzimática , Humanos , Inmunosupresores/farmacología , Activación de Linfocitos/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
2.
Nat Immunol ; 17(6): 704-11, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27064374

RESUMEN

The asymmetric partitioning of fate-determining proteins has been shown to contribute to the generation of CD8(+) effector and memory T cell precursors. Here we demonstrate the asymmetric partitioning of mTORC1 activity after the activation of naive CD8(+) T cells. This results in the generation of two daughter T cells, one of which shows increased mTORC1 activity, increased glycolytic activity and increased expression of effector molecules. The other daughter T cell has relatively low mTORC1 activity and increased lipid metabolism, expresses increased amounts of anti-apoptotic molecules and subsequently displays enhanced long-term survival. Mechanistically, we demonstrate a link between T cell antigen receptor (TCR)-induced asymmetric expression of amino acid transporters and RagC-mediated translocation of mTOR to the lysosomes. Overall, our data provide important insight into how mTORC1-mediated metabolic reprogramming affects the fate decisions of T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , División Celular/inmunología , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Células Precursoras de Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Femenino , Glucólisis , Memoria Inmunológica , Metabolismo de los Lípidos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transporte de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal
3.
Nat Immunol ; 12(4): 295-303, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21358638

RESUMEN

The kinase mTOR has emerged as an important regulator of the differentiation of helper T cells. Here we demonstrate that differentiation into the T(H)1 and T(H)17 subsets of helper T cells was selectively regulated by signaling from mTOR complex 1 (mTORC1) that was dependent on the small GTPase Rheb. Rheb-deficient T cells failed to generate T(H)1 and T(H)17 responses in vitro and in vivo and did not induce classical experimental autoimmune encephalomyelitis (EAE). However, they retained their ability to become T(H)2 cells. Alternatively, when mTORC2 signaling was deleted from T cells, they failed to generate T(H)2 cells in vitro and in vivo but preserved their ability to become T(H)1 and T(H)17 cells. Our data identify mechanisms by which two distinct signaling pathways downstream of mTOR regulate helper cell fate in different ways. These findings define a previously unknown paradigm that links T cell differentiation with selective metabolic signaling pathways.


Asunto(s)
Diferenciación Celular , Proteínas/metabolismo , Transducción de Señal , Linfocitos T Colaboradores-Inductores/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transactivadores/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Citometría de Flujo , Immunoblotting , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Proteínas/genética , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Homóloga de Ras Enriquecida en el Cerebro , Serina-Treonina Quinasas TOR/genética , Células TH1/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo , Transactivadores/genética , Factores de Transcripción
4.
Immunity ; 36(1): 3-5, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22284413

RESUMEN

A hallmark of the adaptive immune response is rapid and robust activation upon rechallenge. In the current issue of Immunity, van der Windt et al. (2012) provide an important link between mitochondrial respiratory capacity and the development of CD8(+) T cell memory.

5.
Immunity ; 34(4): 541-53, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21511183

RESUMEN

The cell-intrinsic mechanisms guiding naive CD8+ T cells for clonal expansion and memory generation via homeostatic proliferation (HP) are unclear. Here, we have shown that HP of naive CD8+ T cells requires IL-7-, but not IL-15-induced mTOR kinase activation. HP-induced mTOR enhances transcription factor T-bet for functional maturation and CD122 expression, which sensitizes for an IL-15-dependent memory transition by favoring transcription factor Eomesodermin over T-bet. Inhibition of mTOR blocks T-bet and CD122 expression but preserves memory in an IL-15-independent manner by promoting Eomesodermin expression. The ability of rapamycin to augment HP-induced memory was cell-intrinsic given that silencing mTOR in CD8+ T cells generated identical outcomes. Strikingly, HP-induced CD8+ T cell memory generated by IL-15-dependent or -independent mechanisms demonstrated identical tumor efficacy. These results indicate a central role for mTOR in HP-induced CD8+ T cell responses and demonstrate the importance for CD8+ memory in HP-induced tumor efficacy.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Homeostasis , Memoria Inmunológica , Neoplasias/inmunología , Serina-Treonina Quinasas TOR/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Progresión de la Enfermedad , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias/patología
6.
Trends Immunol ; 36(1): 13-20, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25522665

RESUMEN

Mammalian/mechanistic target of rapamycin (mTOR) is emerging as an important integrator of environmental cues critical for the regulation of T cell activation, differentiation, and function. Recent studies leveraging pharmacologic inhibition or T cell specific genetic deletion of signaling components in the mTOR pathway have provided important insights into the mechanisms involved, and have been informative in defining targets downstream of mTOR that promote immune regulation. However, these studies have also presented confusing and, at times, contradictory findings, highlighting the complexities involved in examining the mTOR pathway in distinct contexts. Here, we review current understanding of the roles of mTOR in T cell biology, highlighting emerging concepts and areas of investigation where the precise role of mTOR has yet to be fully discerned.


Asunto(s)
Linfocitos T/inmunología , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Eliminación de Gen , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Transducción de Señal , Linfocitos T/citología , Serina-Treonina Quinasas TOR/genética
7.
Proc Natl Acad Sci U S A ; 108(45): 18348-53, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22042850

RESUMEN

Stimulation of resting CD4(+) T lymphocytes leads to rapid proliferation and differentiation into effector (Teff) or inducible regulatory (Treg) subsets with specific functions to promote or suppress immunity. Importantly, Teff and Treg use distinct metabolic programs to support subset specification, survival, and function. Here, we describe that the orphan nuclear receptor estrogen-related receptor-α (ERRα) regulates metabolic pathways critical for Teff. Resting CD4(+) T cells expressed low levels of ERRα protein that increased on activation. ERRα deficiency reduced activated T-cell numbers in vivo and cytokine production in vitro but did not seem to modulate immunity through inhibition of activating signals or viability. Rather, ERRα broadly affected metabolic gene expression and glucose metabolism essential for Teff. In particular, up-regulation of Glut1 protein, glucose uptake, and mitochondrial processes were suppressed in activated ERRα(-/-) T cells and T cells treated with two chemically independent ERRα inhibitors or by shRNAi. Acute ERRα inhibition also blocked T-cell growth and proliferation. This defect appeared as a result of inadequate glucose metabolism, because provision of lipids, but not increased glucose uptake or pyruvate, rescued ATP levels and cell division. Additionally, we have shown that Treg requires lipid oxidation, whereas Teff uses glucose metabolism, and lipid addition selectively restored Treg--but not Teff--generation after acute ERRα inhibition. Furthermore, in vivo inhibition of ERRα reduced T-cell proliferation and Teff generation in both immunization and experimental autoimmune encephalomyelitis models. Thus, ERRα is a selective transcriptional regulator of Teff metabolism that may provide a metabolic means to modulate immunity.


Asunto(s)
Diferenciación Celular , Activación de Linfocitos , Receptores de Estrógenos/fisiología , Linfocitos T/inmunología , Animales , Proliferación Celular , Glucosa/metabolismo , Homeostasis , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Receptores de Estrógenos/genética , Receptor Relacionado con Estrógeno ERRalfa
8.
Hum Mol Genet ; 18(13): 2378-87, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19357198

RESUMEN

Tuberous sclerosis complex (TSC) is a tumor suppressor gene syndrome in which hamartomas develop in multiple organ systems. Knockout and conditional alleles of Tsc1 and Tsc2 have been previously reported. Here, we describe the generation of a novel hypomorphic allele of Tsc2 (del3), in which exon 3, encoding 37 amino acids near the N terminus of tuberin, is deleted. Embryos homozygous for the del3 allele survive until E13.5, 2 days longer than Tsc2 null embryos. Embryos die from underdevelopment of the liver, deficient hematopoiesis, aberrant vascular development and hemorrhage. Mice that are heterozygous for the del3 allele have a markedly reduced kidney tumor burden in comparison with conventional Tsc2(+/-) mice. Murine embryo fibroblast (MEF) cultures that are homozygous for the del3 allele express mutant tuberin at low levels, and show enhanced activation of mTORC1, similar to Tsc2 null MEFs. Furthermore, the mutant cells show prominent reduction in the activation of AKT. Similar findings were made in the analysis of homozygous del3 embryo lysates. Tsc2-del3 demonstrates GTPase activating protein activity comparable to that of wild-type Tsc2 in a functional assay. These findings indicate that the del3 allele is a hypomorphic allele of Tsc2 with partial function due to reduced expression, and highlight the consistency of AKT downregulation when Tsc1/Tsc2 function is reduced. Tsc2-del3 mice also serve as a model for hypomorphic TSC2 missense mutations reported in TSC patients.


Asunto(s)
Modelos Animales de Enfermedad , Hamartoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Alelos , Animales , Línea Celular , Exones , Femenino , Eliminación de Gen , Hamartoma/embriología , Hamartoma/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa
9.
J Neurosci ; 28(21): 5422-32, 2008 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-18495876

RESUMEN

Tuberous sclerosis (TSC) is a hamartoma syndrome attributable to mutations in either TSC1 or TSC2 in which brain involvement causes epilepsy, mental retardation, and autism. We have reported recently (Meikle et al., 2007) a mouse neuronal model of TSC in which Tsc1 is ablated in most neurons during cortical development. We have tested rapamycin and RAD001 [40-O-(2-hydroxyethyl)-rapamycin], both mammalian target of rapamycin mTORC1 inhibitors, as potential therapeutic agents in this model. Median survival is improved from 33 d to more than 100 d; behavior, phenotype, and weight gain are all also markedly improved. There is brain penetration of both drugs, with accumulation over time with repetitive treatment, and effective reduction of levels of phospho-S6, a downstream target of mTORC1. In addition, there is restoration of phospho-Akt and phospho-glycogen synthase kinase 3 levels in the treated mice, consistent with restoration of Akt function. Neurofilament abnormalities, myelination, and cell enlargement are all improved by the treatment. However, dysplastic neuronal features persist, and there are only modest changes in dendritic spine density and length. Strikingly, mice treated with rapamycin or RAD001 for 23 d only (postnatal days 7-30) displayed a persistent improvement in phenotype, with median survival of 78 d. In summary, rapamycin/RAD001 are highly effective therapies for this neuronal model of TSC, with benefit apparently attributable to effects on mTORC1 and Akt signaling and, consequently, cell size and myelination. Although caution is appropriate, the results suggest the possibility that rapamycin/RAD001 may have benefit in the treatment of TSC brain disease, including infantile spasms.


Asunto(s)
Inmunosupresores/uso terapéutico , Proteína Oncogénica v-akt/metabolismo , Transducción de Señal/fisiología , Sirolimus/uso terapéutico , Factores de Transcripción/metabolismo , Esclerosis Tuberosa , Factores de Edad , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Tamaño de la Célula/efectos de los fármacos , Dendritas/efectos de los fármacos , Dendritas/patología , Modelos Animales de Enfermedad , Everolimus , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/patología , Transducción de Señal/efectos de los fármacos , Sirolimus/análogos & derivados , Factores de Tiempo , Esclerosis Tuberosa/tratamiento farmacológico , Esclerosis Tuberosa/mortalidad , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
10.
Mol Cancer ; 8: 38, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19527517

RESUMEN

BACKGROUND: Tuberous sclerosis (TSC) is a hamartoma syndrome in which renal and lung tumors cause the greatest morbidity. Loss of either TSC1 or TSC2 in TSC hamartomas leads to activation of mTORC1 and suppression of AKT. Recent studies indicate that inhibition of mTORC1 with RAD001 (everolimus) leads to rebound activation of AKT, which could protect tumors from drug-induced cell death. Here we examine the potential benefit of inhibition of both mTOR and AKT signaling in a mouse model of TSC, using a dual pan class I PI3K/mTOR catalytic small molecule inhibitor NVP-BEZ235. RESULTS: Using ENU to enhance Tsc2+- kidney tumor development, both RAD001 (10 mg/kg PO 5 d/week) and NVP-BEZ235 (45 mg/kg PO QD) had equivalent effects in suppressing tumor development during a 4 week treatment period, with a 99% reduction in tumor cell mass. Marked reduction in activation of mTORC1, induction of cell cycle arrest, and absence of apoptotic cell death was seen in mice treated with either drug. However, when either was discontinued, there was prompt recovery of tumor growth, with extensive proliferation. CONCLUSION: Both mTORC1 blockade alone and combined PI3K-mTOR blockade lead to suppression of tumor development but not tumor elimination in this TSC model.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Portadoras/antagonistas & inhibidores , Neoplasias Renales/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Sirolimus/análogos & derivados , Factores de Transcripción/antagonistas & inhibidores , Esclerosis Tuberosa/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Etilnitrosourea , Everolimus , Imidazoles/farmacología , Imidazoles/uso terapéutico , Neoplasias Renales/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejos Multiproteicos , Recurrencia Local de Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas , Quinolinas/farmacología , Quinolinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR , Factores de Transcripción/metabolismo , Esclerosis Tuberosa/patología
11.
J Immunother Cancer ; 7(1): 225, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31439037

RESUMEN

BACKGROUND: The safety, efficacy, pharmacokinetics, and pharmacodynamics of the anti-programmed cell death-1 antibody MEDI0680 were evaluated in a phase I, multicenter, dose-escalation study in advanced solid malignancies. METHODS: MEDI0680 was administered intravenously once every 2 weeks (Q2W) or once every 3 weeks at 0.1, 0.5, 2.5, 10 or 20 mg/kg. Two cohorts received 20 mg/kg once a week for 2 or 4 weeks, then 20 mg/kg Q2W. All were treated for 12 months or until progression. The primary endpoint was safety. Secondary endpoints were efficacy and pharmacokinetics. Exploratory endpoints included pharmacodynamics. RESULTS: Fifty-eight patients were treated. Median age was 62.5 years and 81% were male. Most had kidney cancer (n = 36) or melanoma (n = 9). There were no dose-limiting toxicities. Treatment-related adverse events occurred in 83% and were grade ≥ 3 in 21%. Objective clinical responses occurred in 8/58 patients (14%): 5 with kidney cancer, including 1 with a complete response, and 3 with melanoma. The relationship between dose and serum levels was predictable and linear, with apparent receptor saturation at 10 mg/kg Q2W and all 20 mg/kg cohorts. CONCLUSIONS: MEDI0680 induced peripheral T-cell proliferation and increased plasma IFNγ and associated chemokines regardless of clinical response. CD8+ T-cell tumor infiltration and tumoral gene expression of IFNG, CD8A, CXCL9, and granzyme K (GZMK) were also increased following MEDI0680 administration. TRIAL REGISTRATION: NCT02013804 ; date of registration December 12, 2013.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Melanoma/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
12.
J Neurosci ; 27(21): 5546-58, 2007 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-17522300

RESUMEN

Tuberous sclerosis (TSC) is a hamartoma syndrome caused by mutations in TSC1 or TSC2 in which cerebral cortical tubers and seizures are major clinical issues. We have engineered mice in which most cortical neurons lose Tsc1 expression during embryonic development. These Tsc1 mutant mice display several neurological abnormalities beginning at postnatal day 5 with subsequent failure to thrive and median survival of 35 d. The mice also display clinical and electrographic seizures both spontaneously and with physical stimulation, and some seizures end in a fatal tonic phase. Many cortical and hippocampal neurons are enlarged and/or dysplastic in the Tsc1 mutant mice, strongly express phospho-S6, and are ectopic in multiple sites in the cortex and hippocampus. There is a striking delay in myelination in the mutant mice, which appears to be caused by an inductive neuronal defect. This new TSC brain model replicates several features of human TSC brain lesions and implicates an important function of Tsc1/Tsc2 in neuronal development.


Asunto(s)
Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Neuronas/patología , Convulsiones/patología , Esclerosis Tuberosa/patología , Proteínas Supresoras de Tumor/deficiencia , Animales , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Neuronas/metabolismo , Convulsiones/genética , Convulsiones/metabolismo , Tasa de Supervivencia , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/mortalidad , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
13.
PLoS One ; 10(4): e0121710, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25849206

RESUMEN

mTOR is a central integrator of metabolic and immunological stimuli, dictating immune cell activation, proliferation and differentiation. In this study, we demonstrate that within a clonal population of activated T cells, there exist both mTORhi and mTORlo cells exhibiting highly divergent metabolic and immunologic functions. By taking advantage of the role of mTOR activation in controlling cellular size, we demonstrate that upon antigen recognition, mTORhi CD4+ T cells are destined to become highly glycolytic effector cells. Conversely, mTORlo T cells preferentially develop into long-lived cells that express high levels of Bcl-2, CD25, and CD62L. Furthermore, mTORlo T cells have a greater propensity to differentiate into suppressive Foxp3+ T regulatory cells, and this paradigm was also observed in human CD4+ T cells. Overall, these studies provide the opportunity to track the development of effector and memory T cells from naïve precursors, as well as facilitate the interrogation of immunologic and metabolic programs that inform these fates.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Tamaño de la Célula , Regulación de la Expresión Génica/inmunología , Serina-Treonina Quinasas TOR/inmunología , Animales , Femenino , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Selectina L/inmunología , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-2/inmunología
14.
J Clin Invest ; 125(5): 2090-108, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25893604

RESUMEN

Activation of mTOR-dependent pathways regulates the specification and differentiation of CD4+ T effector cell subsets. Herein, we show that mTOR complex 1 (mTORC1) and mTORC2 have distinct roles in the generation of CD8+ T cell effector and memory populations. Evaluation of mice with a T cell-specific deletion of the gene encoding the negative regulator of mTORC1, tuberous sclerosis complex 2 (TSC2), resulted in the generation of highly glycolytic and potent effector CD8+ T cells; however, due to constitutive mTORC1 activation, these cells retained a terminally differentiated effector phenotype and were incapable of transitioning into a memory state. In contrast, CD8+ T cells deficient in mTORC1 activity due to loss of RAS homolog enriched in brain (RHEB) failed to differentiate into effector cells but retained memory characteristics, such as surface marker expression, a lower metabolic rate, and increased longevity. However, these RHEB-deficient memory-like T cells failed to generate recall responses as the result of metabolic defects. While mTORC1 influenced CD8+ T cell effector responses, mTORC2 activity regulated CD8+ T cell memory. mTORC2 inhibition resulted in metabolic reprogramming, which enhanced the generation of CD8+ memory cells. Overall, these results define specific roles for mTORC1 and mTORC2 that link metabolism and CD8+ T cell effector and memory generation and suggest that these functions have the potential to be targeted for enhancing vaccine efficacy and antitumor immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfopoyesis/fisiología , Complejos Multiproteicos/fisiología , Serina-Treonina Quinasas TOR/fisiología , Traslado Adoptivo , Animales , Relación CD4-CD8 , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Proteínas Portadoras/genética , Línea Celular Tumoral , Desoxiglucosa/farmacología , Desoxiglucosa/uso terapéutico , Femenino , Genes Reporteros , Glucólisis/efectos de los fármacos , Memoria Inmunológica , Interferón gamma/biosíntesis , Activación de Linfocitos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Proteínas de Unión al GTP Monoméricas/deficiencia , Proteínas de Unión al GTP Monoméricas/genética , Complejos Multiproteicos/deficiencia , Complejos Multiproteicos/genética , Neuropéptidos/deficiencia , Neuropéptidos/genética , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Homóloga de Ras Enriquecida en el Cerebro , Proteínas Recombinantes de Fusión/inmunología , Sirolimus/farmacología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/deficiencia , Serina-Treonina Quinasas TOR/genética , Timoma/inmunología , Timoma/terapia , Transducción Genética , Factor de Necrosis Tumoral alfa/biosíntesis
15.
Nat Rev Immunol ; 14(7): 435-46, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24962260

RESUMEN

Over the past decade, our understanding of T cell activation, differentiation and function has markedly expanded, providing a greater appreciation of the signals and pathways that regulate these processes. It has become clear that evolutionarily conserved pathways that regulate stress responses, metabolism, autophagy and survival have crucial and specific roles in regulating T cell responses. Recent studies suggest that the metabolic pathways involving MYC, hypoxia-inducible factor 1α (HIF1α), AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) are activated upon antigen recognition and that they are required for directing the consequences of T cell receptor engagement. The purpose of this Review is to provide an integrated view of the role of these metabolic pathways and of canonical T cell signalling pathways in regulating the outcome of T cell responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Proteínas Quinasas Activadas por AMP/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Memoria Inmunológica/inmunología , Ratones , Proteínas Proto-Oncogénicas c-myc/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/metabolismo , Serina-Treonina Quinasas TOR/inmunología
16.
Artículo en Inglés | MEDLINE | ID: mdl-24100582

RESUMEN

Current models of T-helper-cell differentiation depict the generation of effector cells from a naïve T cell based on the cytokine environment upon T-cell-receptor engagement. We propose a new model of CD4(+) T-cell activation, differentiation, and function whereby the outcome of antigen recognition is dictated by mTOR activity and the subsequent up-regulation of selective metabolic function. Such a model more readily explains the generation of effector and memory cells including the concept of effector and memory Foxp3(+) regulatory cells.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T/citología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Linaje de la Célula , Factores de Transcripción Forkhead/metabolismo , Humanos , Memoria Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA