Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(11): 1190-1196, 2017 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-29044230

RESUMEN

The prevalence of autoimmune disorders in affluent countries has reached epidemic proportions. Over the past 50 years, a reverse trend between the frequency of infectious diseases and the incidence of autoimmune and allergic diseases led to the so-called 'hygiene hypothesis'. Given the epidemiological evidence and recent experimental data, we propose that this concept should also include metabolic pressure secondary to exposure to excessive daily caloric intake and overnutrition. We discuss how metabolic workload can modulate immunological tolerance and review the molecular mechanisms and the state of the art of the field. We also critically evaluate possibilities for restoring immunological homeostasis under conditions of metabolic pressure.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Homeostasis/inmunología , Hipótesis de la Higiene , Autotolerancia/inmunología , Animales , Humanos , Redes y Vías Metabólicas/inmunología , Modelos Inmunológicos , Fenómenos Fisiológicos de la Nutrición/inmunología
2.
Nat Immunol ; 16(11): 1174-84, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26414764

RESUMEN

Human regulatory T cells (T(reg) cells) that develop from conventional T cells (T(conv) cells) following suboptimal stimulation via the T cell antigen receptor (TCR) (induced T(reg) cells (iT(reg) cells)) express the transcription factor Foxp3, are suppressive, and display an active proliferative and metabolic state. Here we found that the induction and suppressive function of iT(reg) cells tightly depended on glycolysis, which controlled Foxp3 splicing variants containing exon 2 (Foxp3-E2) through the glycolytic enzyme enolase-1. The Foxp3-E2-related suppressive activity of iT(reg) cells was altered in human autoimmune diseases, including multiple sclerosis and type 1 diabetes, and was associated with impaired glycolysis and signaling via interleukin 2. This link between glycolysis and Foxp3-E2 variants via enolase-1 shows a previously unknown mechanism for controlling the induction and function of T(reg) cells in health and in autoimmunity.


Asunto(s)
Factores de Transcripción Forkhead/genética , Glucólisis/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Adulto , Empalme Alternativo , Autoinmunidad , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Linfocitos T CD4-Positivos/clasificación , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Estudios de Casos y Controles , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Exones , Ácidos Grasos/metabolismo , Femenino , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Variación Genética , Humanos , Técnicas In Vitro , Masculino , Metaboloma , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Oxidación-Reducción , Fosfopiruvato Hidratasa/antagonistas & inhibidores , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/clasificación , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Adulto Joven
3.
Immunity ; 47(5): 875-889.e10, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166588

RESUMEN

Migration of activated regulatory T (Treg) cells to inflamed tissue is crucial for their immune-modulatory function. While metabolic reprogramming during Treg cell differentiation has been extensively studied, the bioenergetics of Treg cell trafficking remains undefined. We have investigated the metabolic demands of migrating Treg cells in vitro and in vivo. We show that glycolysis was instrumental for their migration and was initiated by pro-migratory stimuli via a PI3K-mTORC2-mediated pathway culminating in induction of the enzyme glucokinase (GCK). Subsequently, GCK promoted cytoskeletal rearrangements by associating with actin. Treg cells lacking this pathway were functionally suppressive but failed to migrate to skin allografts and inhibit rejection. Similarly, human carriers of a loss-of-function GCK regulatory protein gene-leading to increased GCK activity-had reduced numbers of circulating Treg cells. These cells displayed enhanced migratory activity but similar suppressive function, while conventional T cells were unaffected. Thus, GCK-dependent glycolysis regulates Treg cell migration.


Asunto(s)
Glucoquinasa/fisiología , Glucólisis , Linfocitos T Reguladores/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD28/fisiología , Antígeno CTLA-4/fisiología , Células Cultivadas , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Diana Mecanicista del Complejo 2 de la Rapamicina/fisiología , Ratones , Ratones Endogámicos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología
4.
Immunity ; 44(2): 406-21, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26885861

RESUMEN

Human CD4(+)CD25(hi)Foxp3(+)CD127(-) Treg and CD4(+)CD25(-)Foxp3(-) Tconv cell functions are governed by their metabolic requirements. Here we report a comprehensive comparative analysis between ex vivo human Treg and Tconv cells that comprises analyses of the proteomic networks in subcellular compartments. We identified a dominant proteomic signature at the metabolic level that primarily impacted the highly-tuned balance between glucose and fatty-acid oxidation in the two cell types. Ex vivo Treg cells were highly glycolytic while Tconv cells used predominantly fatty-acid oxidation (FAO). When cultured in vitro, Treg cells engaged both glycolysis and FAO to proliferate, while Tconv cell proliferation mainly relied on glucose metabolism. Our unbiased proteomic analysis provides a molecular picture of the impact of metabolism on ex vivo human Treg versus Tconv cell functions that might be relevant for therapeutic manipulations of these cells.


Asunto(s)
Ácidos Grasos/metabolismo , Glucosa/metabolismo , Glucólisis , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Antígenos CD4/metabolismo , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Oxidación-Reducción , Proteómica , Transcriptoma
5.
Immunology ; 167(4): 451-470, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36043705

RESUMEN

Epigenetics connects genetic and environmental factors: it includes DNA methylation, histone post-translational modifications and the regulation of chromatin accessibility by non-coding RNAs, all of which control constitutive or inducible gene transcription. This plays a key role in harnessing the transcriptional programs of both innate and adaptive immune cells due to its plasticity and environmental-driven nature, piloting myeloid and lymphoid cell fate decisions with no change in their genomic sequence. In particular, epigenetic marks at the site of lineage-specific transcription factors and maintenance of cell type-specific epigenetic modifications, referred to as 'epigenetic memory', dictate cell differentiation, cytokine production and functional capacity following repeated antigenic exposure in memory T cells. Moreover, metabolic and epigenetic reprogramming occurring during a primary innate immune response leads to enhanced responses to secondary challenges, a phenomenon known as 'trained immunity'. Here, we discuss how stable and dynamic epigenetic states control immune cell identity and plasticity in physiological and pathological conditions. Dissecting the regulatory circuits of cell fate determination and maintenance is of paramount importance for understanding the delicate balance between immune cell activation and tolerance, in healthy conditions and in autoimmune diseases.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Histonas/metabolismo , Diferenciación Celular/genética , Inmunidad , Inmunidad Innata
7.
Proc Natl Acad Sci U S A ; 116(31): 15625-15634, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31308239

RESUMEN

Chronic obstructive pulmonary disease (COPD) is an inflammatory condition associated with abnormal immune responses, leading to airflow obstruction. Lungs of COPD subjects show accumulation of proinflammatory T helper (Th) 1 and Th17 cells resembling that of autoreactive immune responses. As regulatory T (Treg) cells play a central role in the control of autoimmune responses and their generation and function are controlled by the adipocytokine leptin, we herein investigated the association among systemic leptin overproduction, reduced engagement of glycolysis in T cells, and reduced peripheral frequency of Treg cells in different COPD stages. These phenomena were also associated with an impaired capacity to generate inducible Treg (iTreg) cells from conventional T (Tconv) cells. At the molecular level, we found that leptin inhibited the expression of forkhead-boxP3 (FoxP3) and its splicing variants containing the exon 2 (FoxP3-E2) that correlated inversely with inflammation and weakened lung function during COPD progression. Our data reveal that the immunometabolic pathomechanism leading to COPD progression is characterized by leptin overproduction, a decline in the expression of FoxP3 splicing forms, and an impairment in Treg cell generation and function. These results have potential implications for better understanding the autoimmune-like nature of COPD and the pathogenic events leading to lung damage.


Asunto(s)
Empalme Alternativo/inmunología , Factores de Transcripción Forkhead , Leptina , Enfermedad Pulmonar Obstructiva Crónica , Linfocitos T Reguladores , Femenino , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/inmunología , Humanos , Leptina/biosíntesis , Leptina/inmunología , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/patología , Células Th17/inmunología , Células Th17/metabolismo , Células Th17/patología
8.
J Immunol ; 203(7): 1753-1765, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31462505

RESUMEN

Chronic neuroinflammation is a key pathological hallmark of multiple sclerosis (MS) that suggests that resolution of inflammation by specialized proresolving molecules is dysregulated in the disease. Annexin A1 (ANXA1) is a protein induced by glucocorticoids that facilitates resolution of inflammation through several mechanisms that include an inhibition of leukocyte recruitment and activation. In this study, we investigated the ability of ANXA1 to influence T cell effector function in relapsing/remitting MS (RRMS), an autoimmune disease sustained by proinflammatory Th1/Th17 cells. Circulating expression levels of ANXA1 in naive-to-treatment RRMS subjects inversely correlated with disease score and progression. At the cellular level, there was an impaired ANXA1 production by CD4+CD25- conventional T and CD4+RORγt+ T (Th17) cells from RRMS subjects that associated with an increased migratory capacity in an in vitro model of blood brain barrier. Mechanistically, ANXA1 impaired monocyte maturation secondarily to STAT3 hyperactivation and potently reduced T cell activation, proliferation, and glycolysis. Together, these findings identify impaired disease resolution pathways in RRMS caused by dysregulated ANXA1 expression that could represent new potential therapeutic targets in RRMS.


Asunto(s)
Anexina A1/inmunología , Regulación de la Expresión Génica/inmunología , Activación de Linfocitos , Esclerosis Múltiple/inmunología , Células TH1/inmunología , Células Th17/inmunología , Adulto , Proliferación Celular , Femenino , Glucólisis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/patología , Esclerosis Múltiple/patología , Factor de Transcripción STAT3/inmunología , Índice de Severidad de la Enfermedad , Células TH1/patología , Células Th17/patología
9.
Mult Scler ; 26(10): 1237-1246, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31161863

RESUMEN

BACKGROUND: Previous studies evidenced a link between metabolic dysregulation, inflammation, and neurodegeneration in multiple sclerosis (MS). OBJECTIVES: To explore whether increased adipocyte mass expressed as body mass index (BMI) and increased serum lipids influence cerebrospinal fluid (CSF) inflammation and disease severity. METHODS: In this cross-sectional study, 140 consecutive relapsing-remitting (RR)-MS patients underwent clinical assessment, BMI evaluation, magnetic resonance imaging scan, and blood and CSF collection before any specific drug treatment. The CSF levels of the following cytokines, adipocytokines, and inflammatory factors were measured: interleukin (IL)-6, IL-13, granulocyte macrophage colony-stimulating factor, leptin, ghrelin, osteoprotegerin, osteopontin, plasminogen activator inhibitor-1, resistin, and Annexin A1. Serum levels of triglycerides, total cholesterol (TC), and high-density lipoprotein cholesterol (HDL-C) were assessed. RESULTS: A positive correlation emerged between BMI and Expanded Disability Status Scale score. Obese RR-MS patients showed higher clinical disability, increased CSF levels of the proinflammatory molecules IL-6 and leptin, and reduced concentrations of the anti-inflammatory cytokine IL-13. Moreover, both the serum levels of triglycerides and TC/HDL-C ratio showed a positive correlation with IL-6 CSF concentrations. CONCLUSION: Obesity and altered lipid profile are associated with exacerbated central inflammation and higher clinical disability in RR-MS at the time of diagnosis. Increased adipocytokines and lipids can mediate the negative impact of high adiposity on RR-MS course.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente , Esclerosis Múltiple , Estudios Transversales , Humanos , Inflamación , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple Recurrente-Remitente/complicaciones , Obesidad/complicaciones
11.
Semin Immunol ; 28(5): 491-504, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27720234

RESUMEN

The filed that links immunity and metabolism is rapidly expanding. The adipose tissue, by secreting a series of immune regulators called adipokines, represents the common mediator linking metabolic processes and immune system functions. The dysregulation of adipokine secretion, occurring in obese individuals or in conditions of malnutrition or dietary restriction, affects the activity of immune cells resulting in inflammatory autoimmune responses or increased susceptibility to infectious diseases. Alterations of cell metabolism that characterize several autoimmune diseases strongly support the idea that the immune tolerance is also regulated by metabolic pathways. The comprehension of the molecular mechanisms underlying these alterations may lead to the development of novel therapeutic strategies to control immune cell differentiation and function in conditions of autoimmunity.


Asunto(s)
Autoinmunidad , Metabolismo Energético , Tolerancia Inmunológica , Inmunidad , Adipoquinas/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Dieta , Susceptibilidad a Enfermedades , Humanos , Inmunomodulación , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Desnutrición/inmunología , Desnutrición/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Sobrepeso/inmunología , Sobrepeso/metabolismo
12.
Immunity ; 33(6): 929-41, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21145759

RESUMEN

There is a discrepancy between the in vitro anergic state of CD4(+)CD25(hi)FoxP3(+) regulatory T (Treg) cells and their in vivo proliferative capability. The underlying mechanism of this paradox is unknown. Here we show that the anergic state of Treg cells depends on the elevated activity of the mammalian target of rapamycin (mTOR) pathway induced by leptin: a transient inhibition of mTOR with rapamycin, before T cell receptor (TCR) stimulation, made Treg cells highly proliferative in the absence of exogenous interleukin-2 (IL-2). This was a dynamic and oscillatory phenomenon characterized by an early downregulation of the leptin-mTOR pathway followed by an increase in mTOR activation necessary for Treg cell expansion to occur. These data suggest that energy metabolism, through the leptin-mTOR-axis, sets responsiveness of Treg cells that use this information to control immune tolerance and autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Leptina/metabolismo , Linfocitos T Reguladores/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antígenos CD4/biosíntesis , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Anergia Clonal/efectos de los fármacos , Anergia Clonal/genética , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Factores de Transcripción Forkhead/biosíntesis , Humanos , Interleucina-2/inmunología , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Leptina/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Sirolimus/farmacología , Sirolimus/uso terapéutico , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/inmunología
13.
J Immunol ; 198(10): 3803-3808, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28389590

RESUMEN

Glycogen storage disease type 1b (GSD-1b) is an autosomal-recessive disease caused by mutation of glucose-6-phosphate transporter and characterized by altered glycogen/glucose homeostasis. A higher frequency of autoimmune diseases has been observed in GSD-1b patients, but the molecular determinants leading to this phenomenon remain unknown. To address this question, we investigated the effect of glucose-6-phosphate transporter mutation on immune cell homeostasis and CD4+ T cell functions. In GSD-1b subjects, we found lymphopenia and a reduced capacity of T cells to engage glycolysis upon TCR stimulation. These phenomena associated with reduced expression of the FOXP3 transcription factor, lower suppressive function in peripheral CD4+CD25+FOXP3+ regulatory T cells, and an impaired capacity of CD4+CD25- conventional T cells to induce expression of FOXP3 after suboptimal TCR stimulation. These data unveil the metabolic determinant leading to an increased autoimmunity risk in GSD-1b patients.


Asunto(s)
Autoinmunidad , Enfermedad del Almacenamiento de Glucógeno Tipo I/inmunología , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Glucólisis , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Antiportadores/genética , Antiportadores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Niño , Preescolar , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/fisiopatología , Homeostasis , Humanos , Lactante , Linfopenia/inmunología , Linfopenia/fisiopatología , Masculino , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Mutación , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Adulto Joven
14.
Semin Immunol ; 27(5): 300-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26527507

RESUMEN

The immune system is a highly integrated network of cells sensitive to a number of environmental factors. Interestingly, recent years have seen a dramatic increase in our understanding of how diet makes a crucial contribution to human health, affecting the immune system, secretion of adipocytokines and metabolic pathways. Recent experimental evidence indicates that diet and its components are able to profoundly influence immune responses, thus affecting the development of inflammatory and autoimmune diseases. This review aims to discuss some of the main topics concerning the impact of nutrients and their relative composition on immune cell development and function that may be particularly important for regulating the balance between inflammatory and tolerogenic processes. We also highlight the effects of diet on commensal bacteria and how changes in the composition of the microbiota alter intestinal and systemic immune homeostasis. Finally, we summarize the effects of dietary compounds on epigenetic mechanisms involved in the regulation of several immune related genes.


Asunto(s)
Dieta , Alimentos , Inmunidad , Animales , Metabolismo Energético/inmunología , Epigénesis Genética , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Obesidad/genética , Obesidad/inmunología , Obesidad/metabolismo , Transcripción Genética
15.
J Immunol ; 197(7): 2567-75, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27638939

RESUMEN

Intracellular metabolism is central to cell activity and function. CD4(+)CD25(+) regulatory T cells (Tregs) that express the transcription factor FOXP3 play a pivotal role in the maintenance of immune tolerance to self. Recent studies showed that the metabolism and function of Tregs are influenced significantly by local environmental conditions and the availability of certain metabolites. It also was reported that defined metabolic programs associate with Treg differentiation, expression of FOXP3, and phenotype stabilization. This article reviews how metabolism modulates FOXP3 expression and Treg function, what environmental factors are involved, and how metabolic manipulation could alter Treg frequency and function in physiopathologic conditions.


Asunto(s)
Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos
16.
J Biol Chem ; 291(14): 7221-8, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26887954

RESUMEN

The immune system has evolved to protect multicellular organisms from the attack of a variety of pathogens. To exert this function efficiently, the system has developed the capacity to coordinate the function of different cell types and the ability to down-modulate the response when the foreign attack is over. For decades, immunologists believed that these two characteristics were primarily related to cytokine/chemokine-based communication and cell-to-cell direct contact. More recently, it has been shown that immune cells also communicate by transferring regulatory RNAs, microRNAs in particular, from one cell to the other. Several studies have suggested a functional role of extracellular regulatory RNAs in cell-to-cell communication in different cellular contexts. This minireview focuses on the potential role of extracellular RNA transfer in the regulation of adaptive immune response, also contextualizing it in a broader field of what is known of cell-free RNAs in communication among different organisms in the evolutionary scale.


Asunto(s)
Inmunidad Adaptativa/fisiología , Comunicación Celular/inmunología , MicroARNs/inmunología , Animales , Humanos
17.
Clin Immunol ; 183: 249-253, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28823971

RESUMEN

Reliable immunologic biomarkers able to monitor disease course during multiple sclerosis (MS) are still missing. We aimed at identifying possible immunometabolic biomarkers able to predict the clinical outcome in MS patients during treatment with interferon (IFN)-beta-1a. We measured in 45 relapsing-remitting (RR) MS patients, blood circulating levels of several immunometabolic markers, at enrolment, and correlated their levels to disease activity and progression over time. Higher levels of interleukin (IL)-6, soluble-CD40-ligand (sCD40L) and leptin at baseline associated with a higher relapse rate and a greater risk of experiencing at least one relapse in the following year. Higher values of soluble tumor necrosis factor receptor (sTNF-R) and leptin at baseline were predictive of a higher number of lesions in the following one-year of follow up. In conclusion, our data suggest that an immunometabolic profiling measuring IL-6, sCD40L, leptin and sTNF-R at baseline, could represent a useful tool to predict disease course in RRMS patients during treatment with IFN-beta-1a.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Interferón beta-1a/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Transcriptoma/inmunología , Biomarcadores/sangre , Humanos , Esclerosis Múltiple Recurrente-Remitente/sangre , Valor Predictivo de las Pruebas
18.
Proc Natl Acad Sci U S A ; 110(15): 6193-8, 2013 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-23530205

RESUMEN

Whole-body energy metabolism is regulated by the hypothalamus and has an impact on diverse tissue functions. Here we show that selective knockdown of Sirtuin 1 Sirt1 in hypothalamic Agouti-related peptide-expressing neurons, which renders these cells less responsive to cues of low energy availability, significantly promotes CD4(+) T-cell activation by increasing production of T helper 1 and 17 proinflammatory cytokines via mediation of the sympathetic nervous system. These phenomena were associated with an impaired thymic generation of forkhead box P3 (FoxP3(+)) naturally occurring regulatory T cells and their reduced suppressive capacity in the periphery, which resulted in increased delayed-type hypersensitivity responses and autoimmune disease susceptibility in mice. These observations unmask a previously unsuspected role of hypothalamic feeding circuits in the regulation of adaptive immune response.


Asunto(s)
Hambre , Hipotálamo/patología , Neuronas/patología , Linfocitos T Reguladores/citología , Inmunidad Adaptativa , Alelos , Animales , Antígenos/metabolismo , Autoinmunidad , Dominio Catalítico , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Privación de Alimentos , Factores de Transcripción Forkhead/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Hipotálamo/metabolismo , Inflamación , Ratones , Ratones Noqueados , Ratones Transgénicos , Vaina de Mielina/metabolismo , Sirtuina 1/metabolismo , Timo/metabolismo
19.
Immunol Rev ; 249(1): 116-34, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22889219

RESUMEN

The study of how different intracellular metabolic signaling pathways impact the control of self-immune tolerance and how metabolic dysregulation in overweight, obesity, and diabetes is able to alter self-immune tolerance are topics of intensive investigation. Recent evidence suggests that metabolic and autoimmune diseases, both characterized by chronic inflammation and an altered self-immune tolerance, are more common in affluent countries. The reasons for such phenomena are still not completely understood, but the 'metabolic pressure' induced by nutritional overload, typical of more developed countries, seems to play a role. In this context, the discovery of the adipose tissue-derived hormone leptin has shed fundamental insights on how these processes might occur. We believe that there is a strong relationship among leptin, metabolic state, and immunological self-tolerance. We hypothesize that the leptin-induced metabolic pressure sets the basis for an exaggerated immuno-inflammatory response to altered self or non-self, leading to chronic inflammation, metabolic dysregulation, and autoimmunity in subjects with risk factors (i.e. genetic predisposition, environment, sex, infectious agents, etc). Capitalizing on our joint effort and trans-disciplinary expertise in metabolism, self-tolerance, and autoimmune diseases, this review highlights key questions on the basic mechanisms governing immune tolerance in the context of metabolic and autoimmune disease susceptibility.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Diabetes Mellitus/inmunología , Leptina/metabolismo , Autotolerancia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Animales , Peso Corporal , Diabetes Mellitus/metabolismo , Metabolismo Energético , Humanos , Tolerancia Inmunológica , Inflamación/inmunología , Inflamación/metabolismo , Transducción de Señal
20.
Trends Immunol ; 33(1): 1-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22075206

RESUMEN

Disorders such as obesity and type 2 diabetes have been linked to immune dysfunction, raising the possibility that metabolic alterations can be induced by or be a consequence of alterations in immunological tolerance. Here, we describe how intracellular metabolic signalling pathways can 'sense' host energy/nutritional status, and in response, modulate regulatory T (Treg) cell function. In particular, we focus on mammalian target of rapamycin (mTOR) signalling, and how stimuli such as nutrients and leptin activate mTOR in an oscillatory manner to determine Treg cell proliferation status. We propose that metabolic changes such as nutritional deprivation or overload could dictate the characteristics of the Treg cell compartment and subsequent downstream immune reactions.


Asunto(s)
Tolerancia Inmunológica , Espacio Intracelular/metabolismo , Animales , Homeostasis , Humanos , Espacio Intracelular/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA