Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 669456, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34163475

RESUMEN

In Type 1 Diabetes (T1D), CD4+ T cells initiate autoimmune attack of pancreatic islet ß cells. Importantly, bioenergetic programs dictate T cell function, with specific pathways required for progression through the T cell lifecycle. During activation, CD4+ T cells undergo metabolic reprogramming to the less efficient aerobic glycolysis, similarly to highly proliferative cancer cells. In an effort to limit tumor growth in cancer, use of glycolytic inhibitors have been successfully employed in preclinical and clinical studies. This strategy has also been utilized to suppress T cell responses in autoimmune diseases like Systemic Lupus Erythematosus (SLE), Multiple Sclerosis (MS), and Rheumatoid Arthritis (RA). However, modulating T cell metabolism in the context of T1D has remained an understudied therapeutic opportunity. In this study, we utilized the small molecule PFK15, a competitive inhibitor of the rate limiting glycolysis enzyme 6-phosphofructo-2-kinase/fructose-2,6- biphosphatase 3 (PFKFB3). Our results confirmed PFK15 inhibited glycolysis utilization by diabetogenic CD4+ T cells and reduced T cell responses to ß cell antigen in vitro. In an adoptive transfer model of T1D, PFK15 treatment delayed diabetes onset, with 57% of animals remaining euglycemic at the end of the study period. Protection was due to induction of a hyporesponsive T cell phenotype, characterized by increased and sustained expression of the checkpoint molecules PD-1 and LAG-3 and downstream functional and metabolic exhaustion. Glycolysis inhibition terminally exhausted diabetogenic CD4+ T cells, which was irreversible through restimulation or checkpoint blockade in vitro and in vivo. In sum, our results demonstrate a novel therapeutic strategy to control aberrant T cell responses by exploiting the metabolic reprogramming of these cells during T1D. Moreover, the data presented here highlight a key role for nutrient availability in fueling T cell function and has implications in our understanding of T cell biology in chronic infection, cancer, and autoimmunity.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Glucólisis/efectos de los fármacos , Fosfofructoquinasa-2/antagonistas & inhibidores , Piridinas/farmacología , Quinolinas/farmacología , Traslado Adoptivo , Animales , Antígenos CD/metabolismo , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos NOD , Ratones SCID , Fosfofructoquinasa-2/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Factores de Tiempo , Proteína del Gen 3 de Activación de Linfocitos
2.
Diabetes ; 70(7): 1508-1518, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33906911

RESUMEN

In contrast to the skin and the gut, where somatic stem cells and their niche are well characterized, a definitive pancreatic multipotent cell population in the adult pancreas has yet to be revealed. Of particular interest is whether such cells may be endogenous in patients with diabetes, and if so, can they be used for therapeutic purposes? In the current study, we used two separate reporter lines to target Cre-recombinase expression to the Lgr5- or glucagon-expressing cells in the pancreas. We provide evidence for the existence of a population of cells within and in the proximity of the ducts that transiently express the stem-cell marker Lgr5 during late gestational stages. Careful timing of tamoxifen treatment in Lgr5EGFP-IRES-CreERT2 ;R26 Tomato mice allowed us to show that these Lgr5-expressing progenitor cells can differentiate into α-cells during pregnancy. Furthermore, we report on a spontaneous lineage conversion of α- to ß-cells specifically after parturition. The contribution of Lgr5 progeny to the ß-cell compartment through an α-cell intermediate phase early after pregnancy appears to be part of a novel mechanism that would counterbalance against excessive ß-cell mass reduction during ß-cell involution.


Asunto(s)
Linaje de la Célula , Células Secretoras de Glucagón/citología , Células Secretoras de Insulina/citología , Páncreas/citología , Periodo Posparto/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Células Madre/citología , Animales , Apoptosis , Diferenciación Celular , Femenino , Ratones , Ratones Endogámicos C57BL
3.
Cell Rep ; 27(1): 129-141.e4, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30943396

RESUMEN

Lymphocyte activation gene-3 (LAG-3) is an inhibitory receptor expressed by CD4+ T cells and tempers their homeostatic expansion. Because CD4+ T cell proliferation is tightly coupled to bioenergetics, we investigate the role of LAG-3 in modulating naive CD4+ T cell metabolism. LAG-3 deficiency enhances the metabolic profile of naive CD4+ T cells by elevating levels of mitochondrial biogenesis. In vivo, LAG-3 blockade partially restores expansion and the metabolic phenotype of wild-type CD4+ T cells to levels of Lag3-/- CD4+ T cells, solidifying that LAG-3 controls these processes. Lag3-/- CD4+ T cells also demonstrate greater signal transducer and activator of transcription 5 (STAT5) activation, enabling resistance to interleukin-7 (IL-7) deprivation. These results implicate this pathway as a target of LAG-3-mediated inhibition. Additionally, enhancement of STAT5 activation, as a result of LAG-3 deficiency, contributes to greater activation potential in these cells. These results identify an additional mode of regulation elicited by LAG-3 in controlling CD4+ T cell responses.


Asunto(s)
Antígenos CD/fisiología , Linfocitos T CD4-Positivos , Metabolismo Energético/genética , Mitocondrias/fisiología , Biogénesis de Organelos , Animales , Antígenos CD/genética , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/ultraestructura , Células Cultivadas , Femenino , Activación de Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Fase de Descanso del Ciclo Celular/genética , Proteína del Gen 3 de Activación de Linfocitos
4.
PLoS One ; 12(4): e0175549, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28426686

RESUMEN

The immune system is necessary for protecting against various pathogens. However, under certain circumstances, self-reactive immune cells can drive autoimmunity, like that exhibited in type 1 diabetes (T1D). CD4+ T cells are major contributors to the immunopathology in T1D, and in order to drive optimal T cell activation, third signal reactive oxygen species (ROS) must be present. However, the role ROS play in mediating this process remains to be further understood. Recently, cellular metabolic programs have been shown to dictate the function and fate of immune cells, including CD4+ T cells. During activation, CD4+ T cells must transition metabolically from oxidative phosphorylation to aerobic glycolysis to support proliferation and effector function. As ROS are capable of modulating cellular metabolism in other models, we sought to understand if blocking ROS also regulates CD4+ T cell activation and effector function by modulating T cell metabolism. To do so, we utilized an ROS scavenging and potent antioxidant manganese metalloporphyrin (MnP). Our results demonstrate that redox modulation during activation regulates the mTOR/AMPK axis by maintaining AMPK activation, resulting in diminished mTOR activation and reduced transition to aerobic glycolysis in diabetogenic splenocytes. These results correlated with decreased Myc and Glut1 upregulation, reduced glucose uptake, and diminished lactate production. In an adoptive transfer model of T1D, animals treated with MnP demonstrated delayed diabetes progression, concurrent with reduced CD4+ T cell activation. Our results demonstrate that ROS are required for driving and sustaining T cell activation-induced metabolic reprogramming, and further support ROS as a target to minimize aberrant immune responses in autoimmunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos , Especies Reactivas de Oxígeno/metabolismo , Aerobiosis , Animales , Proliferación Celular , Glucosa/metabolismo , Glucólisis , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Fosforilación Oxidativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA