Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 39
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Blood Adv ; 8(17): 4633-4646, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39042920

RÉSUMÉ

ABSTRACT: Autologous T-cell-based therapies, such as chimeric antigen receptor (CAR) T-cell therapy, exhibit low success rates in chronic lymphocytic leukemia (CLL) and correlate with a dysfunctional T-cell phenotype observed in patients. Despite various proposed mechanisms of T-cell dysfunction in CLL, the specific CLL-derived factors responsible remain unidentified. This study aimed to investigate the mechanisms through which CLL cells suppress CAR T-cell activation and function. We found that CLL-derived T cells get activated, albeit in a delayed fashion, and specifically that restimulation of CAR T cells in the presence of CLL cells causes impaired cytokine production and reduced proliferation. Notably, coculture of T cells with CD40-activated CLL cells did not lead to T-cell dysfunction, and this required direct cell contact between the CD40-stimulated CLL cells and T cells. Inhibition of kinases involved in the CD40 signaling cascade revealed that the Spare Respiratory Capacity (SRC) kinase inhibitor dasatinib prevented rescue of T-cell function independent of CD40-mediated increased levels of costimulatory and adhesion ligands on CLL cells. Transcriptome profiling of CD40-stimulated CLL cells with or without dasatinib identified widespread differential gene expression. Selecting for surface receptor genes revealed CD40-mediated downregulation of the Sialic acid-binding Ig-like lectin 10 (Siglec-10) ligands CD24 and CD52, which was prevented by dasatinib, suggesting a role for these ligands in functional T-cell suppression in CLL. Indeed, blocking CD24 and/or CD52 markedly reduced CAR T-cell dysfunction upon coculture with resting CLL cells. These results demonstrated that T cells derived from CLL patients can be reinvigorated by manipulating CLL-T-cell interactions. Targeting CD24- and CD52-mediated CLL-T-cell interaction could be a promising therapeutic strategy to enhance T-cell function in CLL.


Sujet(s)
Antigènes CD24 , Antigène CD52 , Leucémie chronique lymphocytaire à cellules B , Lymphocytes T , Humains , Leucémie chronique lymphocytaire à cellules B/métabolisme , Leucémie chronique lymphocytaire à cellules B/immunologie , Leucémie chronique lymphocytaire à cellules B/thérapie , Antigène CD52/métabolisme , Lymphocytes T/métabolisme , Lymphocytes T/immunologie , Antigènes CD24/métabolisme , Activation des lymphocytes/immunologie , Ligands , Récepteurs chimériques pour l'antigène/métabolisme
2.
Leukemia ; 37(3): 606-616, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36658390

RÉSUMÉ

T-cell dysregulation in chronic lymphocytic leukemia (CLL) associates with low response rates to autologous T cell-based therapies. How CLL affects antigen-specific T-cell responses remains largely unknown. We investigated (epi)genetic and functional consequences of antigen-specific T-cell responses in presence of CLL in vitro and in an adoptive-transfer murine model. Already at steady-state, antigen-experienced patient-derived T cells were skewed towards short-lived effector cells (SLEC) at the expense of memory-precursor effector cells (MPEC). Stimulation of these T cells in vitro showed rapid induction of effector genes and suppression of key memory transcription factors only in presence of CLL cells, indicating epigenetic regulation. This was investigated in vivo by following antigen-specific responses of naïve OT-I CD8+ cells to mCMV-OVA in presence/absence of TCL1 B-cell leukemia. Presence of leukemia resulted in increased SLEC formation, with disturbed inflammatory cytokine production. Chromatin and transcriptome profiling revealed strong epigenetic modifications, leading to activation of an effector and silencing of a memory profile through presence of CLL cells. Secondary challenge in vivo confirmed dysfunctional memory responses by antigen-experienced OT-I cells generated in presence of CLL. Altogether, we show that presence of CLL induces a short-lived effector phenotype and impaired memory responses by epigenetic reprogramming during primary responses.


Sujet(s)
Leucémie chronique lymphocytaire à cellules B , Souris , Animaux , Leucémie chronique lymphocytaire à cellules B/génétique , Leucémie chronique lymphocytaire à cellules B/thérapie , Épigenèse génétique , Lymphocytes T CD8+ , Antigènes , Facteurs de transcription/génétique
3.
Blood ; 140(6): 630-643, 2022 08 11.
Article de Anglais | MEDLINE | ID: mdl-35486832

RÉSUMÉ

Altered metabolism is a hallmark of both cell division and cancer. Chronic lymphocytic leukemia (CLL) cells circulate between peripheral blood (PB) and lymph nodes (LNs), where they receive proliferative and prosurvival signals from surrounding cells. However, insight into the metabolism of LN CLL and how this may relate to therapeutic response is lacking. To obtain insight into CLL LN metabolism, we applied a 2-tiered strategy. First, we sampled PB from 8 patients at baseline and after 3-month ibrutinib (IBR) treatment, which forces egress of CLL cells from LNs. Second, we applied in vitro B-cell receptor (BCR) or CD40 stimulation to mimic the LN microenvironment and performed metabolomic and transcriptomic analyses. The combined analyses indicated prominent changes in purine, glucose, and glutamate metabolism occurring in the LNs. CD40 signaling mostly regulated amino acid metabolism, tricarboxylic acid cycle (TCA), and energy production. BCR signaling preferably engaged glucose and glycerol metabolism and several biosynthesis routes. Pathway analyses demonstrated opposite effects of in vitro stimulation vs IBR treatment. In agreement, the metabolic regulator MYC and its target genes were induced after BCR/CD40 stimulation and suppressed by IBR. Next, 13C fluxomics performed on CD40/BCR-stimulated cells confirmed a strong contribution of glutamine as fuel for the TCA cycle, whereas glucose was mainly converted into lactate and ribose-5-phosphate. Finally, inhibition of glutamine import with V9302 attenuated CD40/BCR-induced resistance to venetoclax. Together, these data provide insight into crucial metabolic changes driven by the CLL LN microenvironment. The prominent use of amino acids as fuel for the TCA cycle suggests new therapeutic vulnerabilities.


Sujet(s)
Leucémie chronique lymphocytaire à cellules B , Antigènes CD40 , Glucose/métabolisme , Glutamine/métabolisme , Humains , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Noeuds lymphatiques/anatomopathologie , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Microenvironnement tumoral
4.
J Immunother Cancer ; 8(1)2020 06.
Article de Anglais | MEDLINE | ID: mdl-32581054

RÉSUMÉ

BACKGROUND: Bispecific antibodies are promising new therapeutics in B cell malignancies. Whether they lead to potent T cell activation despite described T cell dysfunction in chronic lymphocytic leukemia (CLL), and are able to effectively target high-risk or venetoclax-resistant samples, is currently unknown. METHODS: CD19+ cell lines or primary (high-risk) CLL were cocultured in vitro with healthy donor (HD) or CLL-derived T cells in the presence of a CD3xCD19 dual affinity retargeting molecule (CD3xCD19 DART). Cell cytotoxicity, T cell activation, proliferation and effector molecule production were analyzed using flow cytometry. RESULTS: Here, we report that a bispecific CD3xCD19 DART mediates efficient killing by HD T cells of CD19+ cell-lines and primary CLL cells, regardless of immunoglobulin heavy chain variable region (IGHV) mutational status TP53 status or chemotherapy, ibrutinib or venetoclax sensitivity. Whereas TCR stimulation of CLL-derived T cells resulted in dysfunctional T cell activation and proliferation, treatment with CD3xCD19 DART led to a similar activation profile in CLL-derived and HD-derived T cells. Consistently, co-culture of CLL derived T cells with JeKo-1 or CLL cells in the presence of CD3xCD19 DART resulted in significant cytotoxicity by both CD4+ and CD8+ T cells. On stimulation of CLL cells with CD40L, CLL cells become resistant to the specific inhibitor of anti-apoptotic Bcl-2 protein venetoclax, due to upregulation of Bcl-2 family members such as Bcl-XL. Nevertheless, CD40L stimulated CLL cells were as efficiently lysed on CD3xCD19 DART treatment as unstimulated CLL cells. Further examination of the mechanism of CD3xCD19 DART mediated killing showed that lysis was dependent on granules, but was independent of BAX/BAK or caspase activity, indicating non-apoptotic cell death. CONCLUSIONS: These data show that CD3xCD19 DART in CLL leads to robust T cell activation and lysis of high-risk venetoclax resistant CLL cells through a non-apoptotic mechanism.


Sujet(s)
Anticorps bispécifiques/pharmacologie , Antigènes CD19/immunologie , Composés hétérocycliques bicycliques/pharmacologie , Antigènes CD3/immunologie , Leucémie chronique lymphocytaire à cellules B/immunologie , Activation des lymphocytes/immunologie , Sulfonamides/pharmacologie , Lymphocytes T/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Lymphocytes T CD8+/immunologie , Études cas-témoins , Cytotoxicité immunologique/immunologie , Femelle , Études de suivi , Humains , Chaines lourdes des immunoglobulines/génétique , Région variable d'immunoglobuline/génétique , Immunothérapie , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Leucémie chronique lymphocytaire à cellules B/thérapie , Mâle , Adulte d'âge moyen , Mutation , Pronostic , Protéine p53 suppresseur de tumeur/génétique
5.
Blood ; 134(1): 44-58, 2019 07 04.
Article de Anglais | MEDLINE | ID: mdl-31076448

RÉSUMÉ

In chronic lymphocytic leukemia (CLL), acquired T-cell dysfunction impedes development of effective immunotherapeutic strategies, through as-yet unresolved mechanisms. We have previously shown that CD8+ T cells in CLL exhibit impaired activation and reduced glucose uptake after stimulation. CD8+ T cells in CLL patients are chronically exposed to leukemic B cells, which potentially impacts metabolic homeostasis resulting in aberrant metabolic reprogramming upon stimulation. Here, we report that resting CD8+ T cells in CLL have reduced intracellular glucose transporter 1 (GLUT1) reserves, and have an altered mitochondrial metabolic profile as displayed by increased mitochondrial respiration, membrane potential, and levels of reactive oxygen species. This coincided with decreased levels of peroxisome proliferator-activated receptor γ coactivator 1-α, and in line with that, CLL-derived CD8+ T cells showed impaired mitochondrial biogenesis upon stimulation. In search of a therapeutic correlate of these findings, we analyzed mitochondrial biogenesis in CD19-directed chimeric antigen receptor (CAR) CD8+ T cells prior to infusion in CLL patients (who were enrolled in NCT01747486 and NCT01029366 [https://clinicaltrials.gov]). Interestingly, in cases with a subsequent complete response, the infused CD8+ CAR T cells had increased mitochondrial mass compared with nonresponders, which positively correlated with the expansion and persistence of CAR T cells. Our findings demonstrate that GLUT1 reserves and mitochondrial fitness of CD8+ T cells are impaired in CLL. Therefore, boosting mitochondrial biogenesis in CAR T cells might improve the efficacy of CAR T-cell therapy and other emerging cellular immunotherapies.


Sujet(s)
Lymphocytes T CD8+/métabolisme , Immunothérapie adoptive , Leucémie chronique lymphocytaire à cellules B/métabolisme , Mitochondries/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Lignée cellulaire tumorale , Femelle , Humains , Leucémie chronique lymphocytaire à cellules B/thérapie , Mâle , Adulte d'âge moyen , Biogenèse des organelles , Récepteurs chimériques pour l'antigène
6.
Cell Metab ; 28(4): 538-540, 2018 10 02.
Article de Anglais | MEDLINE | ID: mdl-30282046

RÉSUMÉ

Previous immunometabolism research using the CPT1 inhibitor etomoxir suggests that long-chain fatty acid oxidation (LC-FAO) supports IL-4-driven alternative macrophage activation (M(IL-4)) and regulates memory and regulatory T cell formation. Divakaruni et al. (2018) and Raud et al. (2018) now report that LC-FAO is largely dispensable for these processes.


Sujet(s)
Acides gras , Macrophages , Composés époxy , Homéostasie , Oxydoréduction
8.
J Immunol ; 197(6): 2532-40, 2016 09 15.
Article de Anglais | MEDLINE | ID: mdl-27511728

RÉSUMÉ

Leukemia can promote T cell dysfunction and exhaustion that contributes to increased susceptibility to infection and mortality. The treatment-independent mechanisms that mediate leukemia-associated T cell impairments are poorly understood, but metabolism tightly regulates T cell function and may contribute. In this study, we show that B cell leukemia causes T cells to become activated and hyporesponsive with increased PD-1 and TIM3 expression similar to exhausted T cells and that T cells from leukemic hosts become metabolically impaired. Metabolic defects included reduced Akt/mammalian target of rapamycin complex 1 (mTORC1) signaling, decreased expression of the glucose transporter Glut1 and hexokinase 2, and reduced glucose uptake. These metabolic changes correlated with increased regulatory T cell frequency and expression of PD-L1 and Gal-9 on both leukemic and stromal cells in the leukemic microenvironment. PD-1, however, was not sufficient to drive T cell impairment, as in vivo and in vitro anti-PD-1 blockade on its own only modestly improved T cell function. Importantly, impaired T cell metabolism directly contributed to dysfunction, as a rescue of T cell metabolism by genetically increasing Akt/mTORC1 signaling or expression of Glut1 partially restored T cell function. Enforced Akt/mTORC1 signaling also decreased expression of inhibitory receptors TIM3 and PD-1, as well as partially improved antileukemia immunity. Similar findings were obtained in T cells from patients with acute or chronic B cell leukemia, which were also metabolically exhausted and had defective Akt/mTORC1 signaling, reduced expression of Glut1 and hexokinase 2, and decreased glucose metabolism. Thus, B cell leukemia-induced inhibition of T cell Akt/mTORC1 signaling and glucose metabolism drives T cell dysfunction.


Sujet(s)
Transporteur de glucose de type 1/antagonistes et inhibiteurs , Glucose/métabolisme , Leucémie chronique lymphocytaire à cellules B/immunologie , Complexes multiprotéiques/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Lymphocytes T/immunologie , Sérine-thréonine kinases TOR/métabolisme , Animaux , Métabolisme glucidique , Lignée cellulaire tumorale , Glucose/antagonistes et inhibiteurs , Transporteur de glucose de type 1/génétique , Glycolyse , Humains , Activation des lymphocytes , Complexe-1 cible mécanistique de la rapamycine , Souris , Rate/cytologie , Rate/immunologie
9.
Cell ; 166(1): 63-76, 2016 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-27293185

RÉSUMÉ

Activated effector T (TE) cells augment anabolic pathways of metabolism, such as aerobic glycolysis, while memory T (TM) cells engage catabolic pathways, like fatty acid oxidation (FAO). However, signals that drive these differences remain unclear. Mitochondria are metabolic organelles that actively transform their ultrastructure. Therefore, we questioned whether mitochondrial dynamics controls T cell metabolism. We show that TE cells have punctate mitochondria, while TM cells maintain fused networks. The fusion protein Opa1 is required for TM, but not TE cells after infection, and enforcing fusion in TE cells imposes TM cell characteristics and enhances antitumor function. Our data suggest that, by altering cristae morphology, fusion in TM cells configures electron transport chain (ETC) complex associations favoring oxidative phosphorylation (OXPHOS) and FAO, while fission in TE cells leads to cristae expansion, reducing ETC efficiency and promoting aerobic glycolysis. Thus, mitochondrial remodeling is a signaling mechanism that instructs T cell metabolic programming.


Sujet(s)
Dynamique mitochondriale , Lymphocytes T/cytologie , Lymphocytes T/métabolisme , Animaux , Différenciation cellulaire , Transport d'électrons , Acides gras/métabolisme , dGTPases/métabolisme , Glycolyse , Humains , Mémoire immunologique , Souris , Souris de lignée C57BL , Oxydoréduction , Transduction du signal , Lymphocytes T/immunologie
10.
Curr Protoc Immunol ; 113: 3.16B.1-3.16B.14, 2016 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-27038461

RÉSUMÉ

This unit contains several protocols to determine the energy utilization of T cells in real-time using a Seahorse Extracellular Flux Analyzer (http://www.seahorsebio.com). The advantages to using this machine over traditional metabolic assays include the simultaneous measurement of glycolysis and mitochondrial respiration, in real-time, on relatively small numbers of cells, without any radioactivity. The Basic Protocol describes a standard mitochondrial stress test on the XF(e) 96, which yields information about oxidative phosphorylation and glycolysis, two energy-generating pathways. The alternate protocols provide examples of adaptations to the Basic Protocol, including adjustments for the use of the XF(e) 24. A protocol for real-time bioenergetic responses to T cell activation allows for the analysis of immediate metabolic changes after T cell receptor stimulation. Specific substrate utilization can be determined by the use of differential assay media, or the injection of drugs that specifically affect certain metabolic processes. Accurate cell numbers, purity, and viability are critical to obtain reliable results.


Sujet(s)
Techniques de biocapteur , Métabolisme énergétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Animaux , Espace extracellulaire/métabolisme , Acides gras/métabolisme , Galactose/métabolisme , Mitochondries/métabolisme , Phosphorylation oxydative , Smegmamorpha , Stress physiologique
11.
Cell ; 162(6): 1229-41, 2015 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-26321679

RÉSUMÉ

Failure of T cells to protect against cancer is thought to result from lack of antigen recognition, chronic activation, and/or suppression by other cells. Using a mouse sarcoma model, we show that glucose consumption by tumors metabolically restricts T cells, leading to their dampened mTOR activity, glycolytic capacity, and IFN-γ production, thereby allowing tumor progression. We show that enhancing glycolysis in an antigenic "regressor" tumor is sufficient to override the protective ability of T cells to control tumor growth. We also show that checkpoint blockade antibodies against CTLA-4, PD-1, and PD-L1, which are used clinically, restore glucose in tumor microenvironment, permitting T cell glycolysis and IFN-γ production. Furthermore, we found that blocking PD-L1 directly on tumors dampens glycolysis by inhibiting mTOR activity and decreasing expression of glycolysis enzymes, reflecting a role for PD-L1 in tumor glucose utilization. Our results establish that tumor-imposed metabolic restrictions can mediate T cell hyporesponsiveness during cancer.


Sujet(s)
Lymphocytes T CD8+/métabolisme , Glycolyse , Lymphocytes TIL/métabolisme , Tumeurs/métabolisme , Microenvironnement tumoral , Animaux , Anticorps monoclonaux/administration et posologie , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/immunologie , Lymphocytes T CD8+/immunologie , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/immunologie , Interféron gamma/immunologie , Lymphocytes TIL/immunologie , Souris , Tumeurs/immunologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie
13.
PLoS Pathog ; 11(7): e1005027, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-26204259

RÉSUMÉ

Impaired immune responses in the elderly lead to reduced vaccine efficacy and increased susceptibility to viral infections. Although several groups have documented age-dependent defects in adaptive immune priming, the deficits that occur prior to antigen encounter remain largely unexplored. Herein, we identify novel mechanisms for compromised adaptive immunity that occurs with aging in the context of infection with West Nile virus (WNV), an encephalitic flavivirus that preferentially causes disease in the elderly. An impaired IgM and IgG response and enhanced vulnerability to WNV infection during aging was linked to delayed germinal center formation in the draining lymph node (DLN). Adoptive transfer studies and two-photon intravital microscopy revealed a decreased trafficking capacity of donor naïve CD4+ T cells from old mice, which manifested as impaired T cell diapedesis at high endothelial venules and reduced cell motility within DLN prior to antigen encounter. Furthermore, leukocyte accumulation in the DLN within the first few days of WNV infection or antigen-adjuvant administration was diminished more generally in old mice and associated with a second aging-related defect in local cytokine and chemokine production. Thus, age-dependent cell-intrinsic and environmental defects in the DLN result in delayed immune cell recruitment and antigen recognition. These deficits compromise priming of early adaptive immune responses and likely contribute to the susceptibility of old animals to acute WNV infection.


Sujet(s)
Immunité acquise/immunologie , Anticorps antiviraux/immunologie , Noeuds lymphatiques/virologie , Fièvre à virus West Nile/virologie , Virus du Nil occidental/isolement et purification , Vieillissement , Animaux , Encéphale/immunologie , Cytokines/métabolisme , Noeuds lymphatiques/immunologie , Souris , Fièvre à virus West Nile/immunologie , Virus du Nil occidental/immunologie
14.
Immunity ; 42(1): 41-54, 2015 Jan 20.
Article de Anglais | MEDLINE | ID: mdl-25607458

RÉSUMÉ

Naive T cells undergo metabolic reprogramming to support the increased energetic and biosynthetic demands of effector T cell function. However, how nutrient availability influences T cell metabolism and function remains poorly understood. Here we report plasticity in effector T cell metabolism in response to changing nutrient availability. Activated T cells were found to possess a glucose-sensitive metabolic checkpoint controlled by the energy sensor AMP-activated protein kinase (AMPK) that regulated mRNA translation and glutamine-dependent mitochondrial metabolism to maintain T cell bioenergetics and viability. T cells lacking AMPKα1 displayed reduced mitochondrial bioenergetics and cellular ATP in response to glucose limitation in vitro or pathogenic challenge in vivo. Finally, we demonstrated that AMPKα1 is essential for T helper 1 (Th1) and Th17 cell development and primary T cell responses to viral and bacterial infections in vivo. Our data highlight AMPK-dependent regulation of metabolic homeostasis as a key regulator of T cell-mediated adaptive immunity.


Sujet(s)
AMP-Activated Protein Kinases/métabolisme , Lymphocytes T CD4+/physiologie , Lymphocytes T CD8+/physiologie , Sous-type H1N1 du virus de la grippe A/immunologie , Infections à Orthomyxoviridae/métabolisme , AMP-Activated Protein Kinases/génétique , Adaptation physiologique/immunologie , Animaux , Cellules cultivées , Reprogrammation cellulaire/génétique , Reprogrammation cellulaire/immunologie , Métabolisme énergétique , Glucose/métabolisme , Glutamine/métabolisme , Humains , Immunomodulation , Activation des lymphocytes/génétique , Métabolomique , Souris , Souris de lignée C57BL , Souris knockout , Infections à Orthomyxoviridae/immunologie , Biosynthèse des protéines/génétique
15.
Immunity ; 41(1): 75-88, 2014 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-25001241

RÉSUMÉ

Generation of CD8(+) memory T cells requires metabolic reprogramming that is characterized by enhanced mitochondrial fatty-acid oxidation (FAO). However, where the fatty acids (FA) that fuel this process come from remains unclear. While CD8(+) memory T cells engage FAO to a greater extent, we found that they acquired substantially fewer long-chain FA from their external environment than CD8(+) effector T (Teff) cells. Rather than using extracellular FA directly, memory T cells used extracellular glucose to support FAO and oxidative phosphorylation (OXPHOS), suggesting that lipids must be synthesized to generate the substrates needed for FAO. We have demonstrated that memory T cells rely on cell intrinsic expression of the lysosomal hydrolase LAL (lysosomal acid lipase) to mobilize FA for FAO and memory T cell development. Our observations link LAL to metabolic reprogramming in lymphocytes and show that cell intrinsic lipolysis is deterministic for memory T cell fate.


Sujet(s)
Lymphocytes T CD8+/immunologie , Acides gras/métabolisme , Mémoire immunologique/immunologie , Lipolyse/immunologie , Sterol Esterase/métabolisme , 4-Butyrolactone/analogues et dérivés , 4-Butyrolactone/pharmacologie , Transfert adoptif , Animaux , Lymphocytes T CD8+/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Fatty acid synthases/antagonistes et inhibiteurs , Fatty acid synthases/génétique , Acides gras/biosynthèse , Glucose/métabolisme , Interleukine-15/immunologie , Interleukine-2/immunologie , Lipolyse/génétique , Activation des lymphocytes/immunologie , Souris , Souris de lignée C57BL , Souris transgéniques , Mitochondries/métabolisme , Oxydoréduction , Phosphorylation oxydative , Oxygène/métabolisme , Protein kinases/génétique , Interférence par ARN , Petit ARN interférent , Sterol Esterase/biosynthèse
16.
Nat Immunol ; 15(4): 323-32, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24562310

RÉSUMÉ

The ligation of Toll-like receptors (TLRs) leads to rapid activation of dendritic cells (DCs). However, the metabolic requirements that support this process remain poorly defined. We found that DC glycolytic flux increased within minutes of exposure to TLR agonists and that this served an essential role in supporting the de novo synthesis of fatty acids for the expansion of the endoplasmic reticulum and Golgi required for the production and secretion of proteins that are integral to DC activation. Signaling via the kinases TBK1, IKKɛ and Akt was essential for the TLR-induced increase in glycolysis by promoting the association of the glycolytic enzyme HK-II with mitochondria. In summary, we identified the rapid induction of glycolysis as an integral component of TLR signaling that is essential for the anabolic demands of the activation and function of DCs.


Sujet(s)
Cellules dendritiques/immunologie , Glycolyse , I-kappa B Kinase/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Lymphocytes T/immunologie , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , Cellules cultivées , Acides gras/biosynthèse , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/génétique , Glycolyse/immunologie , Hexokinase/métabolisme , I-kappa B Kinase/génétique , Lipopolysaccharides/immunologie , Lipopolysaccharides/pharmacologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes c-akt/métabolisme , Petit ARN interférent/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Récepteurs de type Toll/agonistes
17.
Proc Natl Acad Sci U S A ; 110(35): 14336-41, 2013 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-23940348

RÉSUMÉ

A characteristic of memory T (TM) cells is their ability to mount faster and stronger responses to reinfection than naïve T (TN) cells do in response to an initial infection. However, the mechanisms that allow this rapid recall are not completely understood. We found that CD8 TM cells have more mitochondrial mass than CD8 TN cells and, that upon activation, the resulting secondary effector T (TE) cells proliferate more quickly, produce more cytokines, and maintain greater ATP levels than primary effector T cells. We also found that after activation, TM cells increase oxidative phosphorylation and aerobic glycolysis and sustain this increase to a greater extent than TN cells, suggesting that greater mitochondrial mass in TM cells not only promotes oxidative capacity, but also glycolytic capacity. We show that mitochondrial ATP is essential for the rapid induction of glycolysis in response to activation and the initiation of proliferation of both TN and TM cells. We also found that fatty acid oxidation is needed for TM cells to rapidly respond upon restimulation. Finally, we show that dissociation of the glycolysis enzyme hexokinase from mitochondria impairs proliferation and blocks the rapid induction of glycolysis upon T-cell receptor stimulation in TM cells. Our results demonstrate that greater mitochondrial mass endows TM cells with a bioenergetic advantage that underlies their ability to rapidly recall in response to reinfection.


Sujet(s)
Lymphocytes T CD8+ , Mémoire immunologique , Animaux , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Métabolisme énergétique , Activation des lymphocytes , Souris , Taille de la mitochondrie
18.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-23746840

RÉSUMÉ

A "switch" from oxidative phosphorylation (OXPHOS) to aerobic glycolysis is a hallmark of T cell activation and is thought to be required to meet the metabolic demands of proliferation. However, why proliferating cells adopt this less efficient metabolism, especially in an oxygen-replete environment, remains incompletely understood. We show here that aerobic glycolysis is specifically required for effector function in T cells but that this pathway is not necessary for proliferation or survival. When activated T cells are provided with costimulation and growth factors but are blocked from engaging glycolysis, their ability to produce IFN-γ is markedly compromised. This defect is translational and is regulated by the binding of the glycolysis enzyme GAPDH to AU-rich elements within the 3' UTR of IFN-γ mRNA. GAPDH, by engaging/disengaging glycolysis and through fluctuations in its expression, controls effector cytokine production. Thus, aerobic glycolysis is a metabolically regulated signaling mechanism needed to control cellular function.


Sujet(s)
Glycolyse , Activation des lymphocytes , Phosphorylation oxydative , Lymphocytes T/cytologie , Lymphocytes T/métabolisme , Régions 3' non traduites , Animaux , Prolifération cellulaire , Glyceraldehyde 3-phosphate dehydrogenases/métabolisme , Interféron gamma/génétique , Listeria monocytogenes , Infections à Listeria/immunologie , Souris , Souris de lignée C57BL , Biosynthèse des protéines , Lymphocytes T/immunologie
19.
Immunol Rev ; 249(1): 27-42, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22889213

RÉSUMÉ

Clearance or control of pathogens or tumors usually requires T-cell-mediated immunity. As such, understanding the mechanisms that govern the function, maintenance, and persistence of T cells will likely lead to new treatments for controlling disease. During an immune response, T-cell development is marked by striking changes in metabolism. There is a growing appreciation that these metabolic changes underlie the capacity of T cells to perform particular functions, and this has led to a recent focus on the idea that the manipulation of cellular metabolism can be used to shape adaptive immune responses. Although interest in this area has grown in the last few years, a full understanding of the metabolic control of T-cell functions, particularly during an immune response in vivo, is still lacking. In this review, we first provide a basic overview of metabolism in T cells, and then we focus on recent studies providing new or updated insights into the regulation of metabolic pathways and how they underpin T-cell differentiation and memory T-cell development.


Sujet(s)
Métabolisme énergétique , Mémoire immunologique , Activation des lymphocytes , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Animaux , Différenciation cellulaire , Prolifération cellulaire , Humains , Immunité cellulaire , Mitochondries/métabolisme
20.
Blood ; 120(7): 1422-31, 2012 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-22786879

RÉSUMÉ

TLR agonists initiate a rapid activation program in dendritic cells (DCs) that requires support from metabolic and bioenergetic resources. We found previously that TLR signaling promotes aerobic glycolysis and a decline in oxidative phosphorylation (OXHPOS) and that glucose restriction prevents activation and leads to premature cell death. However, it remained unclear why the decrease in OXPHOS occurs under these circumstances. Using real-time metabolic flux analysis, in the present study, we show that mitochondrial activity is lost progressively after activation by TLR agonists in inflammatory blood monocyte-derived DCs that express inducible NO synthase. We found that this is because of inhibition of OXPHOS by NO and that the switch to glycolysis is a survival response that serves to maintain ATP levels when OXPHOS is inhibited. Our data identify NO as a profound metabolic regulator in inflammatory monocyte-derived DCs.


Sujet(s)
Cellules dendritiques/métabolisme , Cellules dendritiques/anatomopathologie , Glycolyse , Inflammation/anatomopathologie , Monoxyde d'azote/biosynthèse , Adénosine triphosphate/métabolisme , Animaux , Mort cellulaire/effets des médicaments et des substances chimiques , Respiration cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/enzymologie , Glycolyse/effets des médicaments et des substances chimiques , Inflammation/enzymologie , Lipopolysaccharides/pharmacologie , Souris , Souris de lignée C57BL , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Modèles immunologiques , Monocytes/anatomopathologie , Nitric oxide synthase type II/métabolisme , Facteurs temps , Récepteurs de type Toll/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE