Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 25(9): 1637-1649, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39060651

RESUMEN

Approximately 25% of cancers are preceded by chronic inflammation that occurs at the site of tumor development. However, whether this multifactorial oncogenic process, which commonly occurs in the intestines, can be initiated by a specific immune cell population is unclear. Here, we show that an intestinal T cell subset, derived from interleukin-17 (IL-17)-producing helper T (TH17) cells, induces the spontaneous transformation of the intestinal epithelium. This subset produces inflammatory cytokines, and its tumorigenic potential is not dependent on IL-17 production but on the transcription factors KLF6 and T-BET and interferon-γ. The development of this cell type is inhibited by transforming growth factor-ß1 (TGFß1) produced by intestinal epithelial cells. TGFß signaling acts on the pretumorigenic TH17 cell subset, preventing its progression to the tumorigenic stage by inhibiting KLF6-dependent T-BET expression. This study therefore identifies an intestinal T cell subset initiating cancer.


Asunto(s)
Mucosa Intestinal , Factor 6 Similar a Kruppel , Proteínas de Dominio T Box , Células Th17 , Animales , Células Th17/inmunología , Ratones , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Factor 6 Similar a Kruppel/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Transducción de Señal/inmunología , Ratones Endogámicos C57BL , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ratones Noqueados , Interferón gamma/metabolismo , Interferón gamma/inmunología , Interleucina-17/metabolismo , Interleucina-17/inmunología , Ratones Transgénicos , Proteínas Proto-Oncogénicas/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Neoplasias Intestinales/metabolismo , Humanos
2.
Mol Oncol ; 18(7): 1695-1718, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38922759

RESUMEN

Tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T cells have demonstrated remarkable success in the treatment of relapsed/refractory melanoma and hematological malignancies, respectively. These treatments have marked a pivotal shift in cancer management. However, as "living drugs," their effectiveness is dependent on their ability to proliferate and persist in patients. Recent studies indicate that the mechanisms regulating these crucial functions, as well as the T cell's differentiation state, are conditioned by metabolic shifts and the distinct utilization of metabolic pathways. These metabolic shifts, conditioned by nutrient availability as well as cell surface expression of metabolite transporters, are coupled to signaling pathways and the epigenetic landscape of the cell, modulating transcriptional, translational, and post-translational profiles. In this review, we discuss the processes underlying the metabolic remodeling of activated T cells, the impact of a tumor metabolic environment on T cell function, and potential metabolic-based strategies to enhance T cell immunotherapy.


Asunto(s)
Receptores Quiméricos de Antígenos , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Animales , Linfocitos T/inmunología , Linfocitos T/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/metabolismo , Neoplasias/patología , Inmunoterapia Adoptiva/métodos
3.
Immunity ; 57(6): 1260-1273.e7, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38744292

RESUMEN

Upon parasitic helminth infection, activated intestinal tuft cells secrete interleukin-25 (IL-25), which initiates a type 2 immune response during which lamina propria type 2 innate lymphoid cells (ILC2s) produce IL-13. This causes epithelial remodeling, including tuft cell hyperplasia, the function of which is unknown. We identified a cholinergic effector function of tuft cells, which are the only epithelial cells that expressed choline acetyltransferase (ChAT). During parasite infection, mice with epithelial-specific deletion of ChAT had increased worm burden, fitness, and fecal egg counts, even though type 2 immune responses were comparable. Mechanistically, IL-13-amplified tuft cells release acetylcholine (ACh) into the gut lumen. Finally, we demonstrated a direct effect of ACh on worms, which reduced their fecundity via helminth-expressed muscarinic ACh receptors. Thus, tuft cells are sentinels in naive mice, and their amplification upon helminth infection provides an additional type 2 immune response effector function.


Asunto(s)
Acetilcolina , Mucosa Intestinal , Animales , Acetilcolina/metabolismo , Ratones , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Colina O-Acetiltransferasa/metabolismo , Interleucina-13/metabolismo , Interleucina-13/inmunología , Ratones Noqueados , Ratones Endogámicos C57BL , Helmintiasis/inmunología , Helmintiasis/parasitología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Inmunidad Innata , Nematospiroides dubius/inmunología , Células en Penacho
4.
J Immunol ; 212(6): 1029-1039, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38284984

RESUMEN

Both identity and plasticity of CD4 T helper (Th) cells are regulated in part by epigenetic mechanisms. However, a method that reliably and readily profiles DNA base modifications is still needed to finely study Th cell differentiation. Cytosine methylation in CpG context (5mCpG) and cytosine hydroxymethylation (5hmCpG) are DNA modifications that identify stable cell phenotypes, but their potential to characterize intermediate cell transitions has not yet been evaluated. To assess transition states in Th cells, we developed a method to profile Th cell identity using Cas9-targeted single-molecule nanopore sequencing. Targeting as few as 10 selected genomic loci, we were able to distinguish major in vitro polarized murine T cell subtypes, as well as intermediate phenotypes, by their native DNA 5mCpG patterns. Moreover, by using off-target sequences, we were able to infer transcription factor activities relevant to each cell subtype. Detection of 5mCpG and 5hmCpG was validated on intestinal Th17 cells escaping transforming growth factor ß control, using single-molecule adaptive sampling. A total of 21 differentially methylated regions mapping to the 10-gene panel were identified in pathogenic Th17 cells relative to their nonpathogenic counterpart. Hence, our data highlight the potential to exploit native DNA methylation profiling to study physiological and pathological transition states of Th cells.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Animales , Ratones , Citosina , ADN/metabolismo , Células Th17/metabolismo
6.
Proc Natl Acad Sci U S A ; 120(25): e2219431120, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37307458

RESUMEN

Gut microbiota imbalance (dysbiosis) is increasingly associated with pathological conditions, both within and outside the gastrointestinal tract. Intestinal Paneth cells are considered to be guardians of the gut microbiota, but the events linking Paneth cell dysfunction with dysbiosis remain unclear. We report a three-step mechanism for dysbiosis initiation. Initial alterations in Paneth cells, as frequently observed in obese and inflammatorybowel diseases patients, cause a mild remodeling of microbiota, with amplification of succinate-producing species. SucnR1-dependent activation of epithelial tuft cells triggers a type 2 immune response that, in turn, aggravates the Paneth cell defaults, promoting dysbiosis and chronic inflammation. We thus reveal a function of tuft cells in promoting dysbiosis following Paneth cell deficiency and an unappreciated essential role of Paneth cells in maintaining a balanced microbiota to prevent inappropriate activation of tuft cells and deleterious dysbiosis. This succinate-tuft cell inflammation circuit may also contribute to the chronic dysbiosis observed in patients.


Asunto(s)
Disbiosis , Membrana Mucosa , Humanos , Inflamación , Células de Paneth , Succinatos , Ácido Succínico
7.
Cancer Res Commun ; 3(6): 1041-1056, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37377608

RESUMEN

Glioblastomas (GBM) are heterogeneous tumors with high metabolic plasticity. Their poor prognosis is linked to the presence of glioblastoma stem cells (GSC), which support resistance to therapy, notably to temozolomide (TMZ). Mesenchymal stem cells (MSC) recruitment to GBM contributes to GSC chemoresistance, by mechanisms still poorly understood. Here, we provide evidence that MSCs transfer mitochondria to GSCs through tunneling nanotubes, which enhances GSCs resistance to TMZ. More precisely, our metabolomics analyses reveal that MSC mitochondria induce GSCs metabolic reprograming, with a nutrient shift from glucose to glutamine, a rewiring of the tricarboxylic acid cycle from glutaminolysis to reductive carboxylation and increase in orotate turnover as well as in pyrimidine and purine synthesis. Metabolomics analysis of GBM patient tissues at relapse after TMZ treatment documents increased concentrations of AMP, CMP, GMP, and UMP nucleotides and thus corroborate our in vitro analyses. Finally, we provide a mechanism whereby mitochondrial transfer from MSCs to GSCs contributes to GBM resistance to TMZ therapy, by demonstrating that inhibition of orotate production by Brequinar (BRQ) restores TMZ sensitivity in GSCs with acquired mitochondria. Altogether, these results identify a mechanism for GBM resistance to TMZ and reveal a metabolic dependency of chemoresistant GBM following the acquisition of exogenous mitochondria, which opens therapeutic perspectives based on synthetic lethality between TMZ and BRQ. Significance: Mitochondria acquired from MSCs enhance the chemoresistance of GBMs. The discovery that they also generate metabolic vulnerability in GSCs paves the way for novel therapeutic approaches.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células Madre Mesenquimatosas , Humanos , Glioblastoma/tratamiento farmacológico , Resistencia a Antineoplásicos , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Temozolomida/farmacología , Mitocondrias , Células Madre Neoplásicas
8.
Blood ; 141(20): 2520-2536, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-36735910

RESUMEN

Metabolic programs contribute to hematopoietic stem and progenitor cell (HSPC) fate, but it is not known whether the metabolic regulation of protein synthesis controls HSPC differentiation. Here, we show that SLC7A1/cationic amino acid transporter 1-dependent arginine uptake and its catabolism to the polyamine spermidine control human erythroid specification of HSPCs via the activation of the eukaryotic translation initiation factor 5A (eIF5A). eIF5A activity is dependent on its hypusination, a posttranslational modification resulting from the conjugation of the aminobutyl moiety of spermidine to lysine. Notably, attenuation of hypusine synthesis in erythroid progenitors, by the inhibition of deoxyhypusine synthase, abrogates erythropoiesis but not myeloid cell differentiation. Proteomic profiling reveals mitochondrial translation to be a critical target of hypusinated eIF5A, and accordingly, progenitors with decreased hypusine activity exhibit diminished oxidative phosphorylation. This affected pathway is critical for eIF5A-regulated erythropoiesis, as interventions augmenting mitochondrial function partially rescue human erythropoiesis under conditions of attenuated hypusination. Levels of mitochondrial ribosomal proteins (RPs) were especially sensitive to the loss of hypusine, and we find that the ineffective erythropoiesis linked to haploinsufficiency of RPS14 in chromosome 5q deletions in myelodysplastic syndrome is associated with a diminished pool of hypusinated eIF5A. Moreover, patients with RPL11-haploinsufficient Diamond-Blackfan anemia as well as CD34+ progenitors with downregulated RPL11 exhibit a markedly decreased hypusination in erythroid progenitors, concomitant with a loss of mitochondrial metabolism. Thus, eIF5A-dependent protein synthesis regulates human erythropoiesis, and our data reveal a novel role for RPs in controlling eIF5A hypusination in HSPCs, synchronizing mitochondrial metabolism with erythroid differentiation.


Asunto(s)
Proteómica , Espermidina , Humanos , Espermidina/metabolismo , Factores de Iniciación de Péptidos/genética , Diferenciación Celular , Factor 5A Eucariótico de Iniciación de Traducción
9.
Elife ; 122023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36656749

RESUMEN

Quantitative differences in signal transduction are to date an understudied feature of tumour heterogeneity. The MAPK Erk pathway, which is activated in a large proportion of human tumours, is a prototypic example of distinct cell fates being driven by signal intensity. We have used primary hepatocyte precursors transformed with different dosages of an oncogenic form of Ras to model subclonal variations in MAPK signalling. Orthotopic allografts of Ras-transformed cells in immunocompromised mice gave rise to fast-growing aggressive tumours, both at the primary location and in the peritoneal cavity. Fluorescent labelling of cells expressing different oncogene levels, and consequently varying levels of MAPK Erk activation, highlighted the selection processes operating at the two sites of tumour growth. Indeed, significantly higher Ras expression was observed in primary as compared to secondary, metastatic sites, despite the apparent evolutionary trade-off of increased apoptotic death in the liver that correlated with high Ras dosage. Analysis of the immune tumour microenvironment at the two locations suggests that fast peritoneal tumour growth in the immunocompromised setting is abrogated in immunocompetent animals due to efficient antigen presentation by peritoneal dendritic cells. Furthermore, our data indicate that, in contrast to the metastatic-like outgrowth, strong MAPK signalling is required in the primary liver tumours to resist elimination by NK (natural killer) cells. Overall, this study describes a quantitative aspect of tumour heterogeneity and points to a potential vulnerability of a subtype of hepatocellular carcinoma as a function of MAPK Erk signalling intensity.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Humanos , Ratones , Carcinoma Hepatocelular/genética , Células Asesinas Naturales , Neoplasias Hepáticas/genética , Sistema de Señalización de MAP Quinasas , Transducción de Señal , Microambiente Tumoral , Proteínas ras/metabolismo
10.
Eur J Immunol ; 52(3): 418-430, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34961940

RESUMEN

Alterations in cell metabolism can shift the differentiation of immune cells toward a regulatory or inflammatory phenotype, thus, opening up new therapeutic opportunities for immune-related diseases. Indeed, growing knowledge on T- cell metabolism has revealed differences in the metabolic programs of suppressive Tregs as compared to inflammatory Th1 and Th17 cells. In addition to Tregs, IL-10-producing regulatory B cells are crucial for maintaining tolerance, inhibiting inflammation, and autoimmunity. Yet, the metabolic networks regulating diverse B-lymphocyte responses are not well known. Here, we show that glutaminase blockade decreased downstream mTOR activation and attenuated IL-10 secretion. Direct suppression of mTOR activity by rapamycin selectively impaired IL-10 production by B cells whereas secretion was restored upon Glycogen synthase kinase 3 (GSK3) inhibition. Mechanistically, we found mTORC1 activation leads to GSK3 inhibition, identifying a key signalling pathway regulating IL-10 secretion by B lymphocytes. Thus, our results identify glutaminolysis and the mTOR/GSK3 signalling axis, as critical regulators of the generation of IL-10 producing B cells with regulatory functions.


Asunto(s)
Linfocitos B Reguladores , Interleucina-10 , Glutamina/metabolismo , Glucógeno Sintasa Quinasa 3 , Interleucina-10/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
11.
Cell Rep ; 37(5): 109911, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731632

RESUMEN

Suppressive regulatory T cell (Treg) differentiation is controlled by diverse immunometabolic signaling pathways and intracellular metabolites. Here we show that cell-permeable α-ketoglutarate (αKG) alters the DNA methylation profile of naive CD4 T cells activated under Treg polarizing conditions, markedly attenuating FoxP3+ Treg differentiation and increasing inflammatory cytokines. Adoptive transfer of these T cells into tumor-bearing mice results in enhanced tumor infiltration, decreased FoxP3 expression, and delayed tumor growth. Mechanistically, αKG leads to an energetic state that is reprogrammed toward a mitochondrial metabolism, with increased oxidative phosphorylation and expression of mitochondrial complex enzymes. Furthermore, carbons from ectopic αKG are directly utilized in the generation of fatty acids, associated with lipidome remodeling and increased triacylglyceride stores. Notably, inhibition of either mitochondrial complex II or DGAT2-mediated triacylglyceride synthesis restores Treg differentiation and decreases the αKG-induced inflammatory phenotype. Thus, we identify a crosstalk between αKG, mitochondrial metabolism and triacylglyceride synthesis that controls Treg fate.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ácidos Cetoglutáricos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Animales , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Diacilglicerol O-Acetiltransferasa/metabolismo , Fibrosarcoma/genética , Fibrosarcoma/inmunología , Fibrosarcoma/metabolismo , Fibrosarcoma/terapia , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Homeostasis , Humanos , Inmunoterapia Adoptiva , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Fenotipo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/metabolismo
12.
Nat Immunol ; 22(11): 1367-1374, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34686862

RESUMEN

Group 2 innate lymphoid cells (ILC2s) represent innate homologs of type 2 helper T cells (TH2) that participate in immune defense and tissue homeostasis through production of type 2 cytokines. While T lymphocytes metabolically adapt to microenvironmental changes, knowledge of human ILC2 metabolism is limited, and its key regulators are unknown. Here, we show that circulating 'naive' ILC2s have an unexpected metabolic profile with a higher level of oxidative phosphorylation (OXPHOS) than natural killer (NK) cells. Accordingly, ILC2s are severely reduced in individuals with mitochondrial disease (MD) and impaired OXPHOS. Metabolomic and nutrient receptor analysis revealed ILC2 uptake of amino acids to sustain OXPHOS at steady state. Following activation with interleukin-33 (IL-33), ILC2s became highly proliferative, relying on glycolysis and mammalian target of rapamycin (mTOR) to produce IL-13 while continuing to fuel OXPHOS with amino acids to maintain cellular fitness and proliferation. Our results suggest that proliferation and function are metabolically uncoupled in human ILC2s, offering new strategies to target ILC2s in disease settings.


Asunto(s)
Proliferación Celular , Citocinas/metabolismo , Metabolismo Energético , Inmunidad Innata , Activación de Linfocitos , Enfermedades Mitocondriales/metabolismo , Células Th2/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Arginina/metabolismo , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Metabolismo Energético/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Interleucina-33/farmacología , Activación de Linfocitos/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/inmunología , Fenotipo , Células Th2/efectos de los fármacos , Células Th2/inmunología
13.
Cancers (Basel) ; 13(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34503088

RESUMEN

CD19-directed CAR T-cells have been remarkably successful in treating patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) and transformed follicular lymphoma (t-FL). In this cohort study, we treated 60 patients with axicabtagene ciloleucel or tisagenlecleucel. Complete and partial metabolic responses (CMR/PMR) were obtained in 40% and 23% of patients, respectively. After 6.9 months of median follow-up, median progression-free survival (mPFS) and overall survival (mOS) were estimated at 3.1 and 12.3 months, respectively. Statistical analyses revealed that CMR, PFS, and OS were all significantly associated with age-adjusted international prognostic index (aaIPI, p < 0.05). T-cell subset phenotypes in the apheresis product tended to correlate with PFS. Within the final product, increased percentages of both CD4 and CD8 CAR+ effector memory cells (p = 0.02 and 0.01) were significantly associated with CMR. Furthermore, higher CMR/PMR rates were observed in patients with a higher maximal in vivo expansion of CAR T-cells (p = 0.05) and lower expression of the LAG3 and Tim3 markers of exhaustion phenotype (p = 0.01 and p = 0.04). Thus, we find that aaIPI at the time of infusion, phenotype of the CAR T product, in vivo CAR T-cell expansion, and low levels of LAG3/Tim3 are associated with the efficacy of CAR T-cell therapy in DLBCL patients.

14.
Cell Rep ; 34(5): 108723, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33535038

RESUMEN

The metabolic changes controlling the stepwise differentiation of hematopoietic stem and progenitor cells (HSPCs) to mature erythrocytes are poorly understood. Here, we show that HSPC development to an erythroid-committed proerythroblast results in augmented glutaminolysis, generating alpha-ketoglutarate (αKG) and driving mitochondrial oxidative phosphorylation (OXPHOS). However, sequential late-stage erythropoiesis is dependent on decreasing αKG-driven OXPHOS, and we find that isocitrate dehydrogenase 1 (IDH1) plays a central role in this process. IDH1 downregulation augments mitochondrial oxidation of αKG and inhibits reticulocyte generation. Furthermore, IDH1 knockdown results in the generation of multinucleated erythroblasts, a morphological abnormality characteristic of myelodysplastic syndrome and congenital dyserythropoietic anemia. We identify vitamin C homeostasis as a critical regulator of ineffective erythropoiesis; oxidized ascorbate increases mitochondrial superoxide and significantly exacerbates the abnormal erythroblast phenotype of IDH1-downregulated progenitors, whereas vitamin C, scavenging reactive oxygen species (ROS) and reprogramming mitochondrial metabolism, rescues erythropoiesis. Thus, an IDH1-vitamin C crosstalk controls terminal steps of human erythroid differentiation.


Asunto(s)
Ácido Ascórbico/metabolismo , Eritropoyesis/genética , Isocitrato Deshidrogenasa/metabolismo , Mitocondrias/metabolismo , Diferenciación Celular , Humanos
15.
Blood Adv ; 5(1): 26-38, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33570622

RESUMEN

Distinct metabolic demands accompany lymphocyte differentiation into short-lived effector and long-lived memory cells. How bioenergetics processes are structured in innate natural killer (NK) cells remains unclear. We demonstrate that circulating human CD56Dim (NKDim) cells have fused mitochondria and enhanced metabolism compared with CD56Br (NKBr) cells. Upon activation, these 2 subsets showed a dichotomous response, with further mitochondrial potentiation in NKBr cells vs paradoxical mitochondrial fission and depolarization in NKDim cells. The latter effect impaired interferon-γ production, but rescue was possible by inhibiting mitochondrial fragmentation, implicating mitochondrial polarization as a central regulator of NK cell function. NKDim cells are heterogeneous, and mitochondrial polarization was associated with enhanced survival and function in mature NKDim cells, including memory-like human cytomegalovirus-dependent CD57+NKG2C+ subsets. In contrast, patients with genetic defects in mitochondrial fusion had a deficiency in adaptive NK cells, which had poor survival in culture. These results support mitochondrial polarization as a central regulator of mature NK cell fitness.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Humanos , Células Asesinas Naturales , Activación de Linfocitos , Mitocondrias
16.
Mol Cell ; 80(4): 555-557, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33217313

RESUMEN

In this issue of Molecular Cell, Byun et al. (2020) find that the dual targeting of glutamine metabolism and the PD-L1 checkpoint inhibitor augments anti-tumor immunity. Mechanistically, decreased glutamine availability attenuated S-glutathionylation of SERCA, resulting in an increase in cytosolic calcium, enhanced NF-κB activity, and upregulation of programmed death-ligand 1.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Antígeno B7-H1/genética , Glutamina , Humanos , FN-kappa B/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética
17.
Nat Metab ; 1(7): 717-730, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-32373781

RESUMEN

The susceptibility of CD4 T cells to human immunodeficiency virus 1 (HIV-1) infection is regulated by glucose and glutamine metabolism, but the relative contributions of these nutrients to infection are not known. Here we show that glutaminolysis is the major pathway fuelling the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) in T-cell receptor-stimulated naïve, as well as memory CD4, subsets and is required for optimal HIV-1 infection. Under conditions of attenuated glutaminolysis, the α-ketoglutarate (α-KG) TCA rescues early steps in infection; exogenous α-KG promotes HIV-1 reverse transcription, rendering both naïve and memory cells more sensitive to infection. Blocking the glycolytic flux of pyruvate to lactate results in altered glucose carbon allocation to TCA and pentose phosphate pathway intermediates, an increase in OXPHOS and augmented HIV-1 reverse transcription. Moreover, HIV-1 infection is significantly higher in CD4 T cells selected on the basis of high mitochondrial biomass and OXPHOS activity. Therefore, the OXPHOS/aerobic glycolysis balance is a major regulator of HIV-1 infection in CD4 T lymphocytes.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Ciclo del Ácido Cítrico , Glucosa/metabolismo , Glutamina/metabolismo , VIH-1/patogenicidad , Linfocitos T CD4-Positivos/inmunología , Humanos , Activación de Linfocitos
18.
Sci Signal ; 10(501)2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29042482

RESUMEN

The polyphenol resveratrol activates the deacetylase Sirt1, resulting in various antioxidant, chemoprotectant, neuroprotective, cardioprotective, and anti-inflammatory properties. We found that at high concentrations of resveratrol, human CD4+ T cells showed defective antigen receptor signaling and arrest at the G1 stage of the cell cycle, whereas at low concentrations, cells were readily activated and exhibited enhanced Sirt1 deacetylase activity. Nevertheless, low-dose resveratrol rapidly stimulated genotoxic stress in the T cells, which resulted in engagement of a DNA damage response pathway that depended on the kinase ATR [ataxia telangiectasia-mutated (ATM) and Rad3-related], but not ATM, and subsequently in premitotic cell cycle arrest. The concomitant activation of p53 was coupled to the expression of gene products that regulate cell metabolism, leading to a metabolic reprogramming that was characterized by decreased glycolysis, increased glutamine consumption, and a shift to oxidative phosphorylation. These alterations in the bioenergetic homeostasis of CD4+ T cells resulted in enhanced effector function, with both naïve and memory CD4+ T cells secreting increased amounts of the inflammatory cytokine interferon-γ. Thus, our data highlight the wide range of metabolic adaptations that CD4+ T lymphocytes undergo in response to genomic stress.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Daño del ADN , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estilbenos/farmacología , Adulto , Antioxidantes/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Citocinas/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Perfilación de la Expresión Génica/métodos , Glucólisis/efectos de los fármacos , Glucólisis/genética , Humanos , Fosforilación Oxidativa/efectos de los fármacos , Fosforilación/efectos de los fármacos , Resveratrol , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
19.
FEBS Lett ; 591(19): 3104-3118, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28901530

RESUMEN

T cells are stimulated by the engagement of antigen, cytokine, pathogen, and hormone receptors. While research performed over many years has focused on deciphering the molecular components of these pathways, recent data underscore the importance of the metabolic environment in conditioning responses to receptor engagement. The ability of T cells to undergo a massive proliferation and cytokine secretion in response to receptor signals requires alterations to their bioenergetic homeostasis, allowing them to meet new energetic and biosynthetic demands. The metabolic reprogramming of activated T cells is regulated not only by changes in intracellular nutrient uptake and utilization but also by nutrient and oxygen concentrations in the extracellular environment. Notably, the extracellular environment can be profoundly altered by pathological conditions such as infections and tumors, thereby perturbing the metabolism and function of antigen-specific T lymphocytes. This review highlights the interplay between diverse metabolic networks and the transcriptional/epigenetic states that condition T-cell differentiation, comparing the metabolic features of T lymphocytes with other immune cells. We further address recent discoveries in the metabolic pathways that govern T-cell function in physiological and pathological conditions.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Epigénesis Genética , Humanos , Metaboloma/genética , Transducción de Señal
20.
Methods Mol Biol ; 1585: 201-216, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28477198

RESUMEN

Encephalitogenic and colitogenic effector T cells have been implicated in the induction of experimental autoimmune encephalomyelitis (EAE) and inflammatory bowel disease (IBD), respectively. Effector functions of Th1 and Th17 cells have been well characterized and described for the induction and development of EAE and IBD; however, the recently identified Th9 cells have also been suggested to play an important role in these autoimmune pathologies. Th9 cells, primarily characterized by their high level of production of IL-9, are not only essential in controlling extracellular pathogens but also contribute to the development of autoimmunity and allergic inflammation. Furthermore, it was also demonstrated that IL-9 promotes Th17 cell-mediated tissue pathology in EAE and it compromises the barrier functions of the gut in IBD. In vivo adoptive transfer of in vitro differentiated Th9 cells induces the development of autoimmune tissue inflammation in EAE and IBD. Here we describe methods for in vitro differentiation of naïve murine CD4+ T cells to generate IL-9-producing Th9 cells and follow their effector functions in EAE and IBD murine models.


Asunto(s)
Autoinmunidad/fisiología , Encefalomielitis Autoinmune Experimental/metabolismo , Animales , Autoinmunidad/genética , Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Interleucina-9/metabolismo , Ratones , Subgrupos de Linfocitos T/metabolismo , Células TH1/metabolismo , Células Th17/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...