Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell ; 166(1): 63-76, 2016 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-27293185

RESUMEN

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.


Asunto(s)
Dinámicas Mitocondriales , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Diferenciación Celular , Transporte de Electrón , Ácidos Grasos/metabolismo , GTP Fosfohidrolasas/metabolismo , Glucólisis , Humanos , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Transducción de Señal , Linfocitos T/inmunología
2.
Cell ; 162(6): 1229-41, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26321679

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Glucólisis , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Interferón gamma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología
3.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23746840

RESUMEN

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.


Asunto(s)
Glucólisis , Activación de Linfocitos , Fosforilación Oxidativa , Linfocitos T/citología , Linfocitos T/metabolismo , Regiones no Traducidas 3' , Animales , Proliferación Celular , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Interferón gamma/genética , Listeria monocytogenes , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas , Linfocitos T/inmunología
4.
Nat Immunol ; 15(4): 323-32, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24562310

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Glucólisis , Quinasa I-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Ácidos Grasos/biosíntesis , Glucólisis/efectos de los fármacos , Glucólisis/genética , Glucólisis/inmunología , Hexoquinasa/metabolismo , Quinasa I-kappa B/genética , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptores Toll-Like/agonistas
5.
Blood ; 140(6): 630-643, 2022 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-35486832

RESUMEN

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.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Antígenos CD40 , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Ganglios Linfáticos/patología , Receptores de Antígenos de Linfocitos B/metabolismo , Microambiente Tumoral
6.
Immunity ; 42(1): 41-54, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25607458

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Adaptación Fisiológica/inmunología , Animales , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Metabolismo Energético , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Inmunomodulación , Activación de Linfocitos/genética , Metabolómica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Biosíntesis de Proteínas/genética
7.
Immunity ; 41(1): 75-88, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25001241

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ácidos Grasos/metabolismo , Memoria Inmunológica/inmunología , Lipólisis/inmunología , Esterol Esterasa/metabolismo , 4-Butirolactona/análogos & derivados , 4-Butirolactona/farmacología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ácido Graso Sintasas/antagonistas & inhibidores , Ácido Graso Sintasas/genética , Ácidos Grasos/biosíntesis , Glucosa/metabolismo , Interleucina-15/inmunología , Interleucina-2/inmunología , Lipólisis/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , Oxígeno/metabolismo , Proteínas Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño , Esterol Esterasa/biosíntesis
9.
Blood ; 134(1): 44-58, 2019 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31076448

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Inmunoterapia Adoptiva , Leucemia Linfocítica Crónica de Células B/metabolismo , Mitocondrias/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/terapia , Masculino , Persona de Mediana Edad , Biogénesis de Organelos , Receptores Quiméricos de Antígenos
10.
Immunity ; 36(1): 68-78, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22206904

RESUMEN

CD8(+) T cells undergo major metabolic changes upon activation, but how metabolism influences the establishment of long-lived memory T cells after infection remains a key question. We have shown here that CD8(+) memory T cells, but not CD8(+) T effector (Teff) cells, possessed substantial mitochondrial spare respiratory capacity (SRC). SRC is the extra capacity available in cells to produce energy in response to increased stress or work and as such is associated with cellular survival. We found that interleukin-15 (IL-15), a cytokine critical for CD8(+) memory T cells, regulated SRC and oxidative metabolism by promoting mitochondrial biogenesis and expression of carnitine palmitoyl transferase (CPT1a), a metabolic enzyme that controls the rate-limiting step to mitochondrial fatty acid oxidation (FAO). These results show how cytokines control the bioenergetic stability of memory T cells after infection by regulating mitochondrial metabolism.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Mitocondrias/metabolismo , Animales , Linfocitos T CD8-positivos/enzimología , Carnitina O-Palmitoiltransferasa/metabolismo , Respiración de la Célula/fisiología , Ácidos Grasos/metabolismo , Citometría de Flujo , Interleucina-15/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/enzimología , Modelos Biológicos , Oxidación-Reducción , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
J Immunol ; 197(6): 2532-40, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27511728

RESUMEN

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.


Asunto(s)
Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Glucosa/metabolismo , Leucemia Linfocítica Crónica de Células B/inmunología , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Línea Celular Tumoral , Glucosa/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/genética , Glucólisis , Humanos , Activación de Linfocitos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Bazo/citología , Bazo/inmunología
12.
PLoS Pathog ; 11(7): e1005027, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26204259

RESUMEN

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.


Asunto(s)
Inmunidad Adaptativa/inmunología , Anticuerpos Antivirales/inmunología , Ganglios Linfáticos/virología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/aislamiento & purificación , Envejecimiento , Animales , Encéfalo/inmunología , Citocinas/metabolismo , Ganglios Linfáticos/inmunología , Ratones , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología
13.
Proc Natl Acad Sci U S A ; 110(35): 14336-41, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23940348

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Metabolismo Energético , Activación de Linfocitos , Ratones , Tamaño Mitocondrial
14.
Immunol Rev ; 249(1): 27-42, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22889213

RESUMEN

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.


Asunto(s)
Metabolismo Energético , Memoria Inmunológica , Activación de Linfocitos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Inmunidad Celular , Mitocondrias/metabolismo
15.
Blood ; 120(7): 1422-31, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22786879

RESUMEN

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.


Asunto(s)
Células Dendríticas/metabolismo , Células Dendríticas/patología , Glucólisis , Inflamación/patología , Óxido Nítrico/biosíntesis , Adenosina Trifosfato/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/enzimología , Glucólisis/efectos de los fármacos , Inflamación/enzimología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Inmunológicos , Monocitos/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factores de Tiempo , Receptores Toll-Like/metabolismo
16.
J Infect Dis ; 205(12): 1849-57, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22492852

RESUMEN

BACKGROUND: Streptococcus pneumoniae is the most common causative organism in community-acquired pneumonia. Pneumococci that try to invade the lower airways are recognized by innate immune cells through pattern recognition receptors, including Toll-like receptors 2, 4, and 9. Interleukin 1 (IL-1) receptor-associated kinase (IRAK)-M is a proximal inhibitor of Toll-like receptor signaling. METHODS: To determine the role of IRAK-M in host defense during pneumococcal pneumonia, IRAK-M- deficient and wild-type mice were intranasally infected with S. pneumoniae. RESULTS: IRAK-M-deficient mice demonstrated a reduced lethality after infection with S. pneumoniae via the airways. Whereas bacterial burdens were similar in IRAK-M-deficient and wild-type mice early (3 hours) after infection, from 24 hours onward the number of pneumococci recovered from lungs and distant body sites were 10-100-fold lower in the former mouse strain. The diminished bacterial growth and dissemination in IRAK-M-deficient mice were preceded by an increased early influx of neutrophils into lung tissue and elevated pulmonary levels of IL-1ß and CXCL1. IRAK-M deficiency did not influence bacterial growth after intravenous administration of S. pneumoniae. CONCLUSIONS: These data suggest that IRAK-M impairs host defense during pneumococcal pneumonia at the primary site of infection at least in part by inhibiting the early immune response.


Asunto(s)
Interacciones Huésped-Patógeno , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/patología , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad , Animales , Carga Bacteriana , Modelos Animales de Enfermedad , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/deficiencia , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL
17.
Leukemia ; 37(3): 606-616, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36658390

RESUMEN

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.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Ratones , Animales , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/terapia , Epigénesis Genética , Linfocitos T CD8-positivos , Antígenos , Factores de Transcripción/genética
18.
Infect Immun ; 80(5): 1853-7, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22331429

RESUMEN

Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is an important cause of community-acquired sepsis in Southeast Asia and northern Australia. An important controller of the immune system is the pleiotropic cytokine transforming growth factor ß (TGF-ß), of which Smad2 and Smad3 are the major signal transducers. In this study, we aimed to characterize TGF-ß expression and function in experimental melioidosis. TGF-ß expression was determined in 33 patients with culture-proven infection with B. pseudomallei and 30 healthy controls. We found that plasma TGF-ß concentrations were strongly elevated during melioidosis. In line with this finding, TGF-ß expression in C57BL/6 mice intranasally inoculated with B. pseudomallei was enhanced as well. To assess the role of TGF-ß, we inhibited TGF-ß using a selective murine TGF-ß antibody. Treatment of mice with anti-TGF-ß antibody resulted in decreased lung Smad2 phosphorylation. TGF-ß blockade appeared to be protective: mice treated with anti-TGF-ß antibody and subsequently infected with B. pseudomallei showed diminished bacterial loads. Moreover, less distant organ injury was observed in anti-TGF-ß treated mice as shown by reduced blood urea nitrogen (BUN) and aspartate transaminase (AST) values. However, anti-TGF-ß treatment did not have an effect on survival. In conclusion, TGF-ß is upregulated during B. pseudomallei infection and plays a limited but proinflammatory role during experimental melioidosis.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Melioidosis/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Humanos , Inflamación/metabolismo , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Sepsis , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/genética
19.
Mol Med ; 18: 1067-75, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22729155

RESUMEN

Pneumonia is a common cause of morbidity and mortality and the most frequent source of sepsis. Bacteria that try to invade normally sterile body sites are recognized by innate immune cells through pattern recognition receptors, among which toll-like receptors (TLRs) feature prominently. Interleukin-1 receptor (IL-1R)-associated kinase (IRAK)-M is a proximal inhibitor of TLR signaling expressed by epithelial cells and macrophages in the lung. To determine the role of IRAK-M in host defense against bacterial pneumonia, IRAK-M-deficient (IRAK-M(-/-)) and normal wild-type (WT) mice were infected intranasally with Klebsiella pneumoniae. IRAK-M mRNA was upregulated in lungs of WT mice with Klebsiella pneumonia, and the absence of IRAK-M resulted in a strongly improved host defense as reflected by reduced bacterial growth in the lungs, diminished dissemination to distant body sites, less peripheral tissue injury and better survival rates. Although IRAK-M(-/-) alveolar macrophages displayed enhanced responsiveness toward intact K. pneumoniae and Klebsiella lipopolysaccharide (LPS) in vitro, IRAK-M(-/-) mice did not show increased cytokine or chemokine levels in their lungs after infection in vivo. The extent of lung inflammation was increased in IRAK-M(-/-) mice shortly after K. pneumoniae infection, as determined by semiquantitative scoring of specific components of the inflammatory response in lung tissue slides. These data indicate that IRAK-M impairs host defense during pneumonia caused by a common gram-negative respiratory pathogen.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/deficiencia , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Neumonía Bacteriana/inmunología , Animales , Movimiento Celular , Quimiocinas/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-6/metabolismo , Infecciones por Klebsiella/complicaciones , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/patología , Klebsiella pneumoniae/crecimiento & desarrollo , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/patología , Fagocitosis , Neumonía/complicaciones , Neumonía/inmunología , Neumonía/microbiología , Neumonía/patología , Neumonía Bacteriana/complicaciones , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA