Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Isr Med Assoc J ; 23(10): 620-624, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34672442

RESUMEN

BACKGROUND: Cystic periventricular leukomalacia (cPVL) is a strong indicator of subsequent motor and developmental impairments in premature infants. There is a paucity of publications on biomarkers of cPVL. OBJECTIVES: To determine C-reactive protein (CRP) levels during the first week of life of preterm infants who later developed cPVL and to identify the association between CRP levels with perinatal factors. METHODS: We retrospectively included infants ≤ 32 weeks gestation and/or birth weights ≤ 1500 grams; 17 with a cranial ultrasound diagnosis of cPVL and 54 with normal ultrasounds. Serum CRP levels were measured during days 1-7 (CRP1-7d) of life and subdivided into two timing groups: days 1-3 (CRP1-3d) and days 4-7 (CRP4-7d). RESULTS: The cPVL group had significantly higher mean CRP4-7d levels compared to controls (12.75 ± 21.2 vs. 2.23 ± 3.1, respectively, P = 0.03), while CRP1-3d levels were similar. CRP1-7d levels were significantly correlated with maximal fraction of inspired oxygen during the first 12 hours of life (FiO2-12h, r = 0.51, P < 0.001]. Additional risk factors were not associated with CRP levels. CONCLUSIONS: Our finding of elevated CRP4-7d levels and later development of cPVL supports earlier studies on the involvement of inflammation in the pathogenesis of cPVL. Whether CRP could serve as a biomarker of cPVL and its correlation with outcomes, awaits further trials. Furthermore, the correlation between FiO2-12h and CRP1-7d levels suggest that hypoxia and/or hyperoxia may serve as a trigger in the activation of inflammation during the first days of life of preterm infants.


Asunto(s)
Encéfalo/diagnóstico por imagen , Proteína C-Reactiva/análisis , Inflamación/sangre , Leucomalacia Periventricular , Biomarcadores/sangre , Diagnóstico Precoz , Femenino , Edad Gestacional , Humanos , Recién Nacido , Recien Nacido Prematuro , Leucomalacia Periventricular/sangre , Leucomalacia Periventricular/diagnóstico , Masculino , Consumo de Oxígeno/inmunología , Medición de Riesgo , Factores de Riesgo , Ultrasonografía/métodos
2.
Life Sci Alliance ; 4(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34548381

RESUMEN

The mechanisms inducing exhaustion of HIV-specific CD8+ T cells are not fully understood. Metabolic programming directly influences T-cell differentiation, effector function, and memory. We evaluated metabolic profiles of ex vivo CD8+ T cells in HIV-infected individuals. The baseline oxygen consumption rate of CD8+ T cells was elevated in all infected individuals and CD8+ T cells were working at maximal respiratory capacity. The baseline glycolysis rate was enhanced only during early untreated HIV and in viral controllers, but glycolytic capacity was conserved at all stages of infection. CD8+ T-cell mTOR activity was found to be reduced. Enhanced glycolysis was crucial for HIV-specific killing of CD8+ T cells. CD8+ T-cell cytoplasmic GAPDH content was reduced in HIV, but less in early infection and viral controllers. Thus, CD8+ T-cell exhaustion in HIV is characterized by reduced glycolytic activity, enhanced OXPHOS demands, dysregulated mTOR, and reduced cytoplasmic GAPDH. These data provide potential metabolic strategies to reverse CD8+ T-cell dysfunction in HIV.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Glucólisis/inmunología , Infecciones por VIH/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Glucólisis/fisiología , Infecciones por VIH/inmunología , VIH-1/patogenicidad , Humanos , Activación de Linfocitos , Fosforilación Oxidativa , Consumo de Oxígeno/inmunología , Consumo de Oxígeno/fisiología , Serina-Treonina Quinasas TOR/metabolismo
3.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806610

RESUMEN

Microglia, the innate immune cells of the CNS, exhibit long-term response changes indicative of innate immune memory (IIM). Our previous studies revealed IIM patterns of microglia with opposing immune phenotypes: trained immunity after a low dose and immune tolerance after a high dose challenge with pathogen-associated molecular patterns (PAMP). Compelling evidence shows that innate immune cells adopt features of IIM via immunometabolic control. However, immunometabolic reprogramming involved in the regulation of IIM in microglia has not been fully addressed. Here, we evaluated the impact of dose-dependent microglial priming with ultra-low (ULP, 1 fg/mL) and high (HP, 100 ng/mL) lipopolysaccharide (LPS) doses on immunometabolic rewiring. Furthermore, we addressed the role of PI3Kγ on immunometabolic control using naïve primary microglia derived from newborn wild-type mice, PI3Kγ-deficient mice and mice carrying a targeted mutation causing loss of lipid kinase activity. We found that ULP-induced IIM triggered an enhancement of oxygen consumption and ATP production. In contrast, HP was followed by suppressed oxygen consumption and glycolytic activity indicative of immune tolerance. PI3Kγ inhibited glycolysis due to modulation of cAMP-dependent pathways. However, no impact of specific PI3Kγ signaling on immunometabolic rewiring due to dose-dependent LPS priming was detected. In conclusion, immunometabolic reprogramming of microglia is involved in IIM in a dose-dependent manner via the glycolytic pathway, oxygen consumption and ATP production: ULP (ultra-low-dose priming) increases it, while HP reduces it.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/inmunología , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Adenosina Trifosfato/inmunología , Animales , Glucólisis/inmunología , Tolerancia Inmunológica/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Consumo de Oxígeno/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Transducción de Señal/inmunología
4.
J Clin Invest ; 130(3): 1491-1505, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31830003

RESUMEN

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex disease with no known cause or mechanism. There is an increasing appreciation for the role of immune and metabolic dysfunction in the disease. ME/CFS has historically presented in outbreaks, often has a flu-like onset, and results in inflammatory symptoms. Patients suffer from severe fatigue and postexertional malaise. There is little known about the metabolism of specific immune cells in patients with ME/CFS. To investigate immune metabolism in ME/CFS, we isolated CD4+ and CD8+ T cells from 53 patients with ME/CFS and 45 healthy controls. We analyzed glycolysis and mitochondrial respiration in resting and activated T cells, along with markers related to cellular metabolism and plasma cytokines. We found that ME/CFS CD8+ T cells had reduced mitochondrial membrane potential compared with those from healthy controls. Both CD4+ and CD8+ T cells from patients with ME/CFS had reduced glycolysis at rest, whereas CD8+ T cells also had reduced glycolysis following activation. Patients with ME/CFS had significant correlations between measures of T cell metabolism and plasma cytokine abundance that differed from correlations seen in healthy control subjects. Our data indicate that patients have impaired T cell metabolism consistent with ongoing immune alterations in ME/CFS that may illuminate the mechanism behind this disease.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Citocinas , Síndrome de Fatiga Crónica , Mitocondrias , Adulto , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Citocinas/sangre , Citocinas/inmunología , Síndrome de Fatiga Crónica/sangre , Síndrome de Fatiga Crónica/inmunología , Síndrome de Fatiga Crónica/patología , Femenino , Glucólisis/inmunología , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/inmunología , Mitocondrias/metabolismo , Mitocondrias/patología , Consumo de Oxígeno/inmunología
5.
Sci Signal ; 12(599)2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530731

RESUMEN

Transforming growth factor-ß (TGF-ß) is produced by tumors, and increased amounts of this cytokine in the tumor microenvironment and serum are associated with poor patient survival. TGF-ß-mediated suppression of antitumor T cell responses contributes to tumor growth and survival. However, TGF-ß also has tumor-suppressive activity; thus, dissecting cell type-specific molecular effects may inform therapeutic strategies targeting this cytokine. Here, using human peripheral and tumor-associated lymphocytes, we investigated how tumor-derived TGF-ß suppresses a key antitumor function of CD4+ T cells, interferon-γ (IFN-γ) production. Suppression required the expression and phosphorylation of Smad proteins in the TGF-ß signaling pathway, but not their nuclear translocation, and depended on oxygen availability, suggesting a metabolic basis for these effects. Smad proteins were detected in the mitochondria of CD4+ T cells, where they were phosphorylated upon treatment with TGF-ß. Phosphorylated Smad proteins were also detected in the mitochondria of isolated tumor-associated lymphocytes. TGF-ß substantially impaired the ATP-coupled respiration of CD4+ T cells and specifically inhibited mitochondrial complex V (ATP synthase) activity. Last, inhibition of ATP synthase alone was sufficient to impair IFN-γ production by CD4+ T cells. These results, which have implications for human antitumor immunity, suggest that TGF-ß targets T cell metabolism directly, thus diminishing T cell function through metabolic paralysis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interferón gamma/inmunología , Mitocondrias/inmunología , Neoplasias/inmunología , Consumo de Oxígeno/inmunología , Factor de Crecimiento Transformador beta/inmunología , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Humanos , Interferón gamma/metabolismo , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/inmunología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación/inmunología , Transducción de Señal/inmunología , Proteínas Smad/inmunología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/inmunología
6.
Eur J Immunol ; 49(12): 2204-2221, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31373677

RESUMEN

Patients with primary progressive (PP) and secondary progressive (SP) forms of multiple sclerosis (MS) exhibit a sustained increase in the number of Th1, T cytotoxic type-1 and Th17 cells in peripheral blood, suggesting that the progressive phase is characterized by a permanent peripheral immune activation. As T cell functionality and activation are strictly connected to their metabolic profile, we investigated the mitochondrial functionality and metabolic changes of T cell subpopulations in a cohort of progressive MS patients. T cells from progressive patients were characterized by low proliferation and increase of terminally differentiated/exhausted cells. T cells from PP patients showed lower Oxygen Consumption Rate and Extracellular Acidification Rate, lower mitochondrial mass, membrane potential and respiration than those of SP patients, a downregulation of transcription factors supporting respiration and higher tendency to shift towards glycolysis upon stimulation. Furthermore, PP effector memory T cells were characterized by higher Glucose transporter -1 levels and a higher expression of glycolytic-supporting genes if compared to SP patients. Overall, our data suggest that profound differences exist in the phenotypic and metabolic features of T cells from PP and SP patients, even though the two clinical phenotypes are considered part of the same disease spectrum.


Asunto(s)
Memoria Inmunológica , Mitocondrias , Esclerosis Múltiple , Consumo de Oxígeno/inmunología , Linfocitos T , Adulto , Anciano , Femenino , Transportador de Glucosa de Tipo 1/inmunología , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Persona de Mediana Edad , Mitocondrias/inmunología , Mitocondrias/metabolismo , Mitocondrias/patología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
7.
Clin Respir J ; 13(9): 583-589, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31310707

RESUMEN

INTRODUCTION AND OBJECTIVE: Moderate exercise performed in normoxia can be immunostimulatory, while strenuous exercise can be immunosuppressive. However, less is known about the effects of exercise under hypoxia on cytokines. The aim of this study was to evaluate the effects of an acute exercise session performed under hypoxia similar to an altitude of 4200 m on cytokine balance. Our hypothesis was that exercise, even of moderate intensity, associated with hypoxia may induce different changes in relation to the normoxic condition. METHODS: Eight healthy male volunteers were exercised on a treadmill for 1 hour at an intensity of 50% VO2peak under normoxic or hypoxic condition (4200 m). Blood samples were collected at rest and immediately 1 hour after the exercise, respectively to determine cytokines, hormones and metabolites. The two-way ANOVA and the Bonferroni post hoc test were used and the significance adopted was P < .05. RESULTS: While IL-2, the IL-2/IL-4 ratio and glutamine decreased under hypoxia, IL-6 and IL-1ra increased. There were increases in the IL-2/IL-4 ratio, IL-6, IL-1ra and IL-10/TNF-α in normoxia. There were no differences in cortisol or glucose. CONCLUSION: Moderate exercise under hypoxia condition changes the Th1/Th2 balance including IL-2, IL-4 and TNF-α cytokines, suggesting a Th2 response after 1 hour rest.


Asunto(s)
Citocinas/sangre , Ejercicio Físico/fisiología , Hipoxia/sangre , Adulto , Mal de Altura/sangre , Prueba de Esfuerzo/métodos , Glutamina/metabolismo , Humanos , Hipoxia/fisiopatología , Inmunomodulación/fisiología , Inflamación/sangre , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Masculino , Consumo de Oxígeno/inmunología , Consumo de Oxígeno/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Cell Biochem ; 120(4): 5551-5557, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30320909

RESUMEN

The purpose of this study was to investigate the impact of a training season (approximately 7 months) on physiological and salivary immune-endocrine markers in master athletes. Nine male master athletes were evaluated at the beginning of the season (M1) and a week after the main official competition at the end of the sports season (M2). The controlled variables included Maximal oxygen consumption, anthropometric, physiological, and salivary immune-endocrine markers. Master athletes presented a reduced percentage of fat mass and increased lean body mass at the end of the season. VO2max values were similar at M1 and M2, while the maximal heart rate and lactate were lower at M2. No differences were observed in Immunoglobulin A and cortisol levels between moments, whereas testosterone levels and the testosterone/cortisol ratio were significantly lower at the end of the season. The results suggest that maintaining regular training throughout life has positive effects on body composition and improves physiological fitness. However, care should be taken to avoid fatigue as indicated by lower testosterone levels at the end of the season.


Asunto(s)
Atletas , Hidrocortisona , Consumo de Oxígeno/inmunología , Rendimiento Físico Funcional , Saliva , Testosterona , Adulto , Femenino , Humanos , Hidrocortisona/inmunología , Hidrocortisona/metabolismo , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Masculino , Saliva/inmunología , Saliva/metabolismo , Testosterona/inmunología , Testosterona/metabolismo
9.
Methods Mol Biol ; 1862: 173-186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30315468

RESUMEN

Metabolic reprograming controlling macrophage activation and function is emerging as new regulatory circuit on shaping immune responses. Generally, lipopolysaccharides (LPS)-induced pro-inflammatory activated macrophages, known as M1 macrophages, display higher glycolysis. In contrast, interleukin-4 (IL-4)-skewed anti-inflammatory activated macrophages, known as M2 macrophages, mainly rely on oxidative phosphorylation for their bioenergetic demands. Emerging evidence reveals that these metabolic preferences further fine-tune macrophage polarization process, including signaling cascades and epigenetic reprogramming. Thus, specific nutrient microenvironments may affect inflammatory responses of macrophages by intervening these metabolic machineries. How to measure the metabolic switch of macrophages both in vitro and in vivo is an important issue for understanding immunometabolic regulations in macrophages. Here, we describe a basic protocol for examining how glutamine metabolism affects macrophage polarization by using the Extracellular Flux (XF(e)96) Analyzer (Seahorse Bioscience), which takes real-time measurements of oxidative phosphorylation and glycolysis. We also present a detailed procedure for detecting the expression of inflammatory genes in polarized macrophages under glutamine-replete or -deprived conditions.


Asunto(s)
Activación de Macrófagos/inmunología , Espectrometría de Masas/métodos , Análisis de Flujos Metabólicos/métodos , Metabolómica/métodos , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Células Cultivadas , Glutamina/metabolismo , Glucólisis/inmunología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Espectrometría de Masas/instrumentación , Análisis de Flujos Metabólicos/instrumentación , Redes y Vías Metabólicas/inmunología , Metabolómica/instrumentación , Ratones , Ratones Endogámicos C57BL , Fosforilación Oxidativa , Consumo de Oxígeno/inmunología , Cultivo Primario de Células/instrumentación , Cultivo Primario de Células/métodos , Organismos Libres de Patógenos Específicos
10.
J Immunol Methods ; 463: 54-60, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30201392

RESUMEN

Dimethyl sulfoxide (DMSO) is a polar organic solvent used in a wide range of biological applications. DMSO is routinely used as a cryoprotectant for long-term cell freezing as well as to dissolve peptides and drugs for immune cell functional assays. Here, human CD4+ T cell activation, cytokine production, proliferation, and metabolism were investigated after stimulation in the presence of 0.01% to 1%, DMSO, representing concentrations commonly used in vitro. Surface expression of the activation markers CD69, CD25 and CD154 after polyclonal activation of CD4+ T cells was inhibited by 0.25% or higher concentrations of DMSO. The frequencies of IL-21+, IL-4+, and IL-22+ CD4+ T cells, following polyclonal activation were variably inhibited by DMSO at concentrations ranging from 0.25% to 1%, whereas IFNγ+ cells were unaffected. CD4+ T cell proliferation after anti-CD3 or antigen stimulation was inhibited by 0.5% DMSO and abolished by 1% DMSO. After polyclonal stimulation, glucose uptake was inhibited in the presence of 1% DMSO, but only minor effects on CD4+ T cell respiration were observed. Consistent with the immune effects, the gene expression of early signaling and activation pathways were inhibited in CD4+ T cells in the presence of 1% DMSO. Our study revealed that DMSO at concentrations generally used for in vitro studies of T cells impacts multiple features of T cell function. Therefore, we urge care when adding DMSO-containing preparations to T cell cultures.


Asunto(s)
Antígenos CD , Linfocitos T CD4-Positivos , Proliferación Celular/efectos de los fármacos , Citocinas , Dimetilsulfóxido/farmacología , Activación de Linfocitos/efectos de los fármacos , Antígenos CD/inmunología , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/inmunología
11.
Respir Res ; 19(1): 163, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30165864

RESUMEN

BACKGROUND: Autoimmune pulmonary alveolar proteinosis (aPAP) is a rare pulmonary disease caused by functional deficiency of granulocyte-macrophage colony-stimulating factor (GM-CSF). GM-CSF therapy in aPAP has been reported effective in some studies. This meta-analyses aimed to evaluate whether GM-CSF therapy, including inhaled and subcutaneous GM-CSF have therapeutic effect in aPAP patients. METHODS: We analyzed 10 studies searched from PubMed, EmBase, Web of Science, Wiley Online Library and Cochrane Collaboration databases to evaluate the pooled effects of GM-CSF treatment in aPAP patients. RESULTS: Ten observational studies involving 115 aPAP patients were included. The pooled analyses of response rate (81%, p < 0.001), relapse rate (22%, p = 0.009), PaO2 (13.76 mmHg, p < 0.001) and P(A-a)O2 (19.44 mmHg, p < 0.001) showed that GM-CSF treatment was effective on aPAP patients. Further analyses showed that inhaled GM-CSF treatment was more effective than subcutaneous GM-CSF therapy, including a higher response rate (89% vs. 71%, p = 0.023), more improvements in PaO2 (21.02 mmHg vs. 8.28 mmHg, p < 0.001) and P(A-a)O2 (19.63 mmHg vs. 9.15 mmHg, p < 0.001). CONCLUSIONS: As two routes of exogenous GM-CSF treatment, inhaled and subcutaneous were both proven to have effect on aPAP patients. Furthermore, inhaled GM-CSF therapy showed a higher response rate, more improvements on PaO2 and P(A-a)O2 than subcutaneous GM-CSF treatment in aPAP patients, suggesting inhaled GM-CSF therapy could have more benefits on aPAP patients. Therefore, GM-CSF therapy, especially inhaled GM-CSF, might be a promising therapeutic option in treating aPAP.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Proteinosis Alveolar Pulmonar/tratamiento farmacológico , Proteinosis Alveolar Pulmonar/inmunología , Administración por Inhalación , Análisis de los Gases de la Sangre/métodos , Humanos , Inyecciones Subcutáneas , Estudios Observacionales como Asunto/métodos , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/inmunología , Proteinosis Alveolar Pulmonar/diagnóstico , Resultado del Tratamiento
12.
Proc Natl Acad Sci U S A ; 115(34): E8017-E8026, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30072431

RESUMEN

T cells critically depend on reprogramming of metabolic signatures to meet the bioenergetic demands during activation and clonal expansion. Here we identify the transcription factor Nur77 as a cell-intrinsic modulator of T cell activation. Nur77-deficient T cells are highly proliferative, and lack of Nur77 is associated with enhanced T cell activation and increased susceptibility for T cell-mediated inflammatory diseases, such as CNS autoimmunity, allergic contact dermatitis and collagen-induced arthritis. Importantly, Nur77 serves as key regulator of energy metabolism in T cells, restricting mitochondrial respiration and glycolysis and controlling switching between different energy pathways. Transcriptional network analysis revealed that Nur77 modulates the expression of metabolic genes, most likely in close interaction with other transcription factors, especially estrogen-related receptor α. In summary, we identify Nur77 as a transcriptional regulator of T cell metabolism, which elevates the threshold for T cell activation and confers protection in different T cell-mediated inflammatory diseases.


Asunto(s)
Autoinmunidad , Activación de Linfocitos , Mitocondrias , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Consumo de Oxígeno/inmunología , Linfocitos T , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Perfilación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/inmunología , Mitocondrias/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/inmunología , Receptores de Estrógenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor Relacionado con Estrógeno ERRalfa
13.
J Appl Physiol (1985) ; 125(2): 586-595, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29648517

RESUMEN

This study investigated immunophenotypic differences in intracellular thiol redox state of peripheral blood mononuclear cells (PBMCs) isolated from trained [ n = 9, means ± SD: age 28 ± 5 yr; (body mass index) BMI 23.2 ± 2.6 kg/m2; V̇o2max (maximal oxygen intake)56.9 ± 6.1 ml·kg-1·min-1] and recreationally active (RA, n = 11, means ± SD: age 27 ± 6 yr; BMI 24.2 ± 3.7 kg/m2; V̇o2max 45.1 ± 6.4 ml·kg-1·min-1) participants before and after a maximal aerobic exercise tolerance test. Blood samples were taken before (Pre), during (sample acquired at 70% maximum heart rate), immediately after (Post + 0), and 15 min postexercise (Post + 15). PBMCs were isolated, and reduced thiol analysis [fluorescein-5 maleimide (F5M)] by immunophenotype [cluster of differentiation (CD)3+, CD4+, and CD8+] was performed using flow cytometry. A significant increase in cellular F5M fluorescence was observed in CD3+ T cells at Post + 0, with changes driven to a greater extent by CD8+ T cells (fold change in both groups CD4: +2.3, CD8: +2.8; P < 0.05). Further analysis revealed a population of highly reduced CD8+ T cells (CD8+T-reduced+) that significantly increased from Pre to Post + 0 in RA participants only (RA: +272 cell/µl, P < 0.05). To understand these results further, CD8+T-reduced+ and CD8+T-reduced- cells were analyzed for immunophenotype in response to the same exercise protocol ( n = 6, means ± SD: age 24 ± 5 yr; BMI 25.7 ± 4.1 kg·m-2; V̇o2max 41.33 ± 7.63 ml·kg-1·min-1). CD8+T-reduced+ had significantly less lymphoid homing potential (chemokine receptor type 7) Post + 0 compared with Pre. This study is the first, to our knowledge, to demonstrate that lymphocyte populations become more reductive in response to acute exercise. NEW & NOTEWORTHY The study presented provides the first evidence to suggest that cytotoxic T cells become transiently reductive in healthy individuals following a single bout of cycling. Detection of these cells was enabled via the use of a flow cytometric assay that incorporates the thiol reactive probe fluorescein-5 maleimide. Using this method, transient reductive stress in viable T cells is permissible and provides the basis for further research in the area of exercise immunology.


Asunto(s)
Ejercicio Físico/fisiología , Estrés Fisiológico/fisiología , Linfocitos T Citotóxicos/fisiología , Adulto , Prueba de Esfuerzo/métodos , Femenino , Citometría de Flujo , Humanos , Recuento de Leucocitos/métodos , Leucocitos/inmunología , Leucocitos/fisiología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/fisiología , Masculino , Consumo de Oxígeno/inmunología , Consumo de Oxígeno/fisiología , Estrés Fisiológico/inmunología , Linfocitos T Citotóxicos/inmunología
14.
Cell Immunol ; 330: 54-59, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29395037

RESUMEN

Macrophages are innate immune cells that provide host defense and have tissue-specific roles in the maintenance of organ homeostasis and integrity. In most cases macrophages keep us healthy but when their balanced response to damage or homeostatic signals is perturbed, they can drive chronic inflammatory responses and pathology. To fulfil their broad range of functions, macrophages adopt a plethora of activation states. Understanding their regulation and phenotypic heterogeneity is crucial because macrophages are critical in many diseases. Consequently, macrophages have emerged as attractive targets for therapy of diseases in which they determine disease outcome, such as cardiovascular disease, cancer and other Western killer diseases. Recent advances in the flourishing field of immunometabolism highlight that the metabolic profile of macrophages directly regulates their activation status and associated functions. In this short review, we summarize how recent research on the metabolic regulation of macrophages has vividly improved our understanding of macrophage activation. Most of our existing knowledge results from in vitro studies with murine bone marrow-derived macrophages which can't fully grasp the complexity of (micro)environmental control of macrophages in tissues. We therefore highlight current weaknesses and missing links in macrophage immunometabolism research and provide future directions to make the step from the well-controlled plastic in vitro cell culture systems to the complex in vivo tissue environment.


Asunto(s)
Metabolismo Energético/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Mitocondrias/inmunología , Animales , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/metabolismo , Homeostasis/inmunología , Humanos , Macrófagos/metabolismo , Mitocondrias/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Consumo de Oxígeno/inmunología
15.
Biosci Rep ; 37(4)2017 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-28798194

RESUMEN

Metabolism is of central importance for T cell survival and differentiation. It is well known that T cells cannot function in the absence of glucose, but it is less clear how they respond to excessive levels of glucose. In the present study, we investigated how increasing levels of glucose affect T-cell-mediated immune responses. We examined the effects of increased levels of glucose on CD8+ T-cell behaviour in vitro by assessing activation and cytokine production, as well as oxygen consumption rate (OCR), extracellular acidification rate (ECAR) and intracellular signalling. In addition, we assessed in vivo proliferation, cytokine production and cytolytic activity of cells in chemically induced diabetic C57BL/6 mice. Elevated levels of glucose in in vitro cultures had modest effects on proliferation and cytokine production, while in vivo hyperglycaemia had no effect on CD8+ T-cell proliferation, interferon γ (IFNγ) production or cytolytic killing.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Experimental/inmunología , Hiperglucemia/inmunología , Activación de Linfocitos , Animales , Linfocitos T CD8-positivos/patología , Diabetes Mellitus Experimental/patología , Hiperglucemia/patología , Interferón gamma/inmunología , Ratones , Consumo de Oxígeno/inmunología
16.
Arthritis Rheumatol ; 68(11): 2728-2739, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27332042

RESUMEN

OBJECTIVE: Antiphospholipid antibodies (aPL) constitute a diagnostic criterion of systemic lupus erythematosus (SLE), and aPL have been functionally linked to liver disease in patients with SLE. Since the mechanistic target of rapamycin (mTOR) is a regulator of oxidative stress, a pathophysiologic process that contributes to the development of aPL, this study was undertaken in a mouse model of SLE to examine the involvement of liver mitochondria in lupus pathogenesis. METHODS: Mitochondria were isolated from lupus-prone MRL/lpr, C57BL/6.lpr, and MRL mice, age-matched autoimmunity-resistant C57BL/6 mice as negative controls, and transaldolase-deficient mice, a strain that exhibits oxidative stress in the liver. Electron transport chain (ETC) activity was assessed using measurements of oxygen consumption. ETC proteins, which are regulators of mitochondrial homeostasis, and the mTOR complexes mTORC1 and mTORC2 were examined by Western blotting. Anticardiolipin (aCL) and anti-ß2 -glycoprotein I (anti-ß2 GPI) autoantibodies were measured by enzyme-linked immunosorbent assay in mice treated with rapamycin or mice treated with a solvent control. RESULTS: Mitochondrial oxygen consumption was increased in the livers of 4-week-old, disease-free MRL/lpr mice relative to age-matched controls. Levels of the mitophagy initiator dynamin-related protein 1 (Drp1) were depleted while the activity of mTORC1 was increased in MRL/lpr mice. In turn, mTORC2 activity was decreased in MRL and MRL/lpr mice. In addition, levels of aCL and anti-ß2 GPI were elevated preceding the development of nephritis in 4-week-old MRL, C57BL/6.lpr, and MRL/lpr mice. Transaldolase-deficient mice showed increased oxygen consumption, depletion of Drp1, activation of mTORC1, and elevated expression of NADH:ubiquinone oxidoreductase core subunit S3 (NDUFS3), a pro-oxidant subunit of ETC complex I, as well as increased production of aCL and anti-ß2 GPI autoantibodies. Treatment with rapamycin selectively blocked mTORC1 activation, NDUFS3 expression, and aPL production both in transaldolase-deficient mice and in lupus-prone mice. CONCLUSION: In lupus-prone mice, mTORC1-dependent mitochondrial dysfunction contributes to the generation of aPL, suggesting that such mechanisms may represent a treatment target in patients with SLE.


Asunto(s)
Anticuerpos Antifosfolípidos/biosíntesis , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Lupus Eritematoso Sistémico/inmunología , Mitocondrias Hepáticas/metabolismo , Complejos Multiproteicos/metabolismo , Estrés Oxidativo/inmunología , Consumo de Oxígeno/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Anticuerpos Anticardiolipina/biosíntesis , Anticuerpos Anticardiolipina/efectos de los fármacos , Anticuerpos Anticardiolipina/inmunología , Anticuerpos Antifosfolípidos/efectos de los fármacos , Anticuerpos Antifosfolípidos/inmunología , Formación de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/inmunología , Western Blotting , Modelos Animales de Enfermedad , Dinaminas/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunosupresores/farmacología , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Noqueados , Mitocondrias Hepáticas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Sirolimus/farmacología , Transaldolasa/genética , beta 2 Glicoproteína I/inmunología
18.
J Immunol ; 196(1): 80-90, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608911

RESUMEN

We have previously shown that CD4(+) T cells from B6.Sle1Sle2.Sle3 lupus mice and patients present a high cellular metabolism, and a treatment combining 2-deoxy-D-glucose, which inhibits glucose metabolism, and metformin, which inhibits oxygen consumption, normalized lupus T cell functions in vitro and reverted disease in mice. We obtained similar results with B6.lpr mice, another model of lupus, and showed that a continuous treatment is required to maintain the beneficial effect of metabolic inhibitors. Further, we investigated the relative roles of glucose oxidation and pyruvate reduction into lactate in this process. Treatments of B6.Sle1Sle2.Sle3 mice with either 2-deoxy-D-glucose or metformin were sufficient to prevent autoimmune activation, whereas their combination was necessary to reverse the process. Treatment of B6.Sle1Sle2.Sle3 mice with dichloroacetate, an inhibitor of lactate production, failed to effectively prevent or reverse autoimmune pathology. In vitro, CD4(+) T cell activation upregulated the expression of genes that favor oxidative phosphorylation. Blocking glucose oxidation inhibited both IFN-γ and IL-17 production, which could not be achieved by blocking pyruvate reduction. Overall, our data show that targeting glucose oxidation is required to prevent or reverse lupus development in mice, which cannot be achieved by simply targeting the pyruvate-lactate conversion.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Glucosa/metabolismo , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos/inmunología , Oxidación-Reducción/efectos de los fármacos , Animales , Autoinmunidad/efectos de los fármacos , Células Cultivadas , Desoxiglucosa/farmacología , Ácido Dicloroacético/farmacología , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/inmunología , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Ácido Láctico/biosíntesis , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación Oxidativa , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/inmunología , Ácido Pirúvico/metabolismo
19.
J Clin Invest ; 125(12): 4334-48, 2015 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-26517695

RESUMEN

Cellular metabolism is increasingly recognized as a controller of immune cell fate and function. MicroRNA-33 (miR-33) regulates cellular lipid metabolism and represses genes involved in cholesterol efflux, HDL biogenesis, and fatty acid oxidation. Here, we determined that miR-33-mediated disruption of the balance of aerobic glycolysis and mitochondrial oxidative phosphorylation instructs macrophage inflammatory polarization and shapes innate and adaptive immune responses. Macrophage-specific Mir33 deletion increased oxidative respiration, enhanced spare respiratory capacity, and induced an M2 macrophage polarization-associated gene profile. Furthermore, miR-33-mediated M2 polarization required miR-33 targeting of the energy sensor AMP-activated protein kinase (AMPK), but not cholesterol efflux. Notably, miR-33 inhibition increased macrophage expression of the retinoic acid-producing enzyme aldehyde dehydrogenase family 1, subfamily A2 (ALDH1A2) and retinal dehydrogenase activity both in vitro and in a mouse model. Consistent with the ability of retinoic acid to foster inducible Tregs, miR-33-depleted macrophages had an enhanced capacity to induce forkhead box P3 (FOXP3) expression in naive CD4(+) T cells. Finally, treatment of hypercholesterolemic mice with miR-33 inhibitors for 8 weeks resulted in accumulation of inflammation-suppressing M2 macrophages and FOXP3(+) Tregs in plaques and reduced atherosclerosis progression. Collectively, these results reveal that miR-33 regulates macrophage inflammation and demonstrate that miR-33 antagonism is atheroprotective, in part, by reducing plaque inflammation by promoting M2 macrophage polarization and Treg induction.


Asunto(s)
Aterosclerosis/inmunología , Regulación de la Expresión Génica/inmunología , Macrófagos Peritoneales/inmunología , MicroARNs/inmunología , Placa Aterosclerótica/inmunología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/inmunología , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/inmunología , Familia de Aldehído Deshidrogenasa 1 , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Glucólisis/genética , Glucólisis/inmunología , Macrófagos Peritoneales/patología , Ratones , Ratones Noqueados , MicroARNs/genética , Consumo de Oxígeno/genética , Consumo de Oxígeno/inmunología , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Retinal-Deshidrogenasa , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
20.
Semin Immunol ; 27(3): 177-83, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25818531

RESUMEN

Inflammatory diseases in mucosal organs as diverse as the lung, liver and intestine inevitably require the intimate interactions between neutrophils and epithelia. The physiologic consequences of such interactions often determine endpoint organ function, and for this reason, much recent interest has developed in identifying mechanisms and novel targets to promote the resolution of mucosal inflammation. Physiologically-relevant in vitro and in vivo model systems have aided in discovery of novel pathways to define basic inflammatory mechanisms and approaches to defining the concepts of inflammatory resolution. Here, we will review the recent literature regarding the contribution of neutrophils to inflammatory resolution, with an emphasis on the role of the tissue microenvironment, endogenous pathways for promoting resolution and the molecular determinants of neutrophil-epithelial cell interactions during ongoing inflammation. These recent studies highlight the dynamic nature of pro-resolving pathways and lend insight into the complexity of treating mucosal inflammation.


Asunto(s)
Células Epiteliales/inmunología , Homeostasis/inmunología , Inflamación/inmunología , Membrana Mucosa/inmunología , Neutrófilos/inmunología , Comunicación Celular/inmunología , Hipoxia de la Célula/inmunología , Movimiento Celular/inmunología , Microambiente Celular/inmunología , Humanos , Membrana Mucosa/citología , Membrana Mucosa/patología , Consumo de Oxígeno/inmunología , Consumo de Oxígeno/fisiología , Nucleósidos de Purina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA