Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell ; 179(7): 1483-1498.e22, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31813625

RESUMEN

Metabolism has been shown to control peripheral immunity, but little is known about its role in central nervous system (CNS) inflammation. Through a combination of proteomic, metabolomic, transcriptomic, and perturbation studies, we found that sphingolipid metabolism in astrocytes triggers the interaction of the C2 domain in cytosolic phospholipase A2 (cPLA2) with the CARD domain in mitochondrial antiviral signaling protein (MAVS), boosting NF-κB-driven transcriptional programs that promote CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis. cPLA2 recruitment to MAVS also disrupts MAVS-hexokinase 2 (HK2) interactions, decreasing HK enzymatic activity and the production of lactate involved in the metabolic support of neurons. Miglustat, a drug used to treat Gaucher and Niemann-Pick disease, suppresses astrocyte pathogenic activities and ameliorates EAE. Collectively, these findings define a novel immunometabolic mechanism that drives pro-inflammatory astrocyte activities, outlines a new role for MAVS in CNS inflammation, and identifies candidate targets for therapeutic intervention.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Fosfolipasas A2 Secretoras/metabolismo , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Hexoquinasa/metabolismo , Humanos , Ácido Láctico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosfolipasas A2 Secretoras/genética
2.
Nature ; 611(7937): 801-809, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36266581

RESUMEN

Genome-wide association studies have identified risk loci linked to inflammatory bowel disease (IBD)1-a complex chronic inflammatory disorder of the gastrointestinal tract. The increasing prevalence of IBD in industrialized countries and the augmented disease risk observed in migrants who move into areas of higher disease prevalence suggest that environmental factors are also important determinants of IBD susceptibility and severity2. However, the identification of environmental factors relevant to IBD and the mechanisms by which they influence disease has been hampered by the lack of platforms for their systematic investigation. Here we describe an integrated systems approach, combining publicly available databases, zebrafish chemical screens, machine learning and mouse preclinical models to identify environmental factors that control intestinal inflammation. This approach established that the herbicide propyzamide increases inflammation in the small and large intestine. Moreover, we show that an AHR-NF-κB-C/EBPß signalling axis operates in T cells and dendritic cells to promote intestinal inflammation, and is targeted by propyzamide. In conclusion, we developed a pipeline for the identification of environmental factors and mechanisms of pathogenesis in IBD and, potentially, other inflammatory diseases.


Asunto(s)
Ambiente , Herbicidas , Inflamación , Enfermedades Inflamatorias del Intestino , Intestinos , Animales , Ratones , Inflamación/inducido químicamente , Inflamación/etiología , Inflamación/inmunología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Pez Cebra , Aprendizaje Automático , Bases de Datos Factuales , Modelos Animales de Enfermedad , Intestinos/efectos de los fármacos , Intestinos/inmunología , Intestinos/metabolismo , Intestinos/patología , FN-kappa B , Proteína beta Potenciadora de Unión a CCAAT , Receptores de Hidrocarburo de Aril , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Herbicidas/efectos adversos
3.
Nature ; 557(7707): 724-728, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29769726

RESUMEN

Microglia and astrocytes modulate inflammation and neurodegeneration in the central nervous system (CNS)1-3. Microglia modulate pro-inflammatory and neurotoxic activities in astrocytes, but the mechanisms involved are not completely understood4,5. Here we report that TGFα and VEGF-B produced by microglia regulate the pathogenic activities of astrocytes in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Microglia-derived TGFα acts via the ErbB1 receptor in astrocytes to limit their pathogenic activities and EAE development. Conversely, microglial VEGF-B triggers FLT-1 signalling in astrocytes and worsens EAE. VEGF-B and TGFα also participate in the microglial control of human astrocytes. Furthermore, expression of TGFα and VEGF-B in CD14+ cells correlates with the multiple sclerosis lesion stage. Finally, metabolites of dietary tryptophan produced by the commensal flora control microglial activation and TGFα and VEGF-B production, modulating the transcriptional program of astrocytes and CNS inflammation through a mechanism mediated by the aryl hydrocarbon receptor. In summary, we identified positive and negative regulators that mediate the microglial control of astrocytes. Moreover, these findings define a pathway through which microbial metabolites limit pathogenic activities of microglia and astrocytes, and suppress CNS inflammation. This pathway may guide new therapies for multiple sclerosis and other neurological disorders.


Asunto(s)
Astrocitos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/microbiología , Microglía/metabolismo , Animales , Astrocitos/patología , Células Cultivadas , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/microbiología , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/prevención & control , Receptores ErbB/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Inflamación/prevención & control , Receptores de Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Receptores de Hidrocarburo de Aril/metabolismo , Simbiosis , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador alfa/metabolismo , Triptófano/deficiencia , Triptófano/metabolismo , Factor B de Crecimiento Endotelial Vascular/biosíntesis , Factor B de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
4.
Proc Natl Acad Sci U S A ; 114(8): 2012-2017, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28167760

RESUMEN

Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the CNS that causes disability in young adults as a result of the irreversible accumulation of neurological deficits. Although there are potent disease-modifying agents for its initial relapsing-remitting phase, these therapies show limited efficacy in secondary progressive MS (SPMS). Thus, there is an unmet clinical need for the identification of disease mechanisms and potential therapeutic approaches for SPMS. Here, we show that the sphingosine 1-phosphate receptor (S1PR) modulator fingolimod (FTY720) ameliorated chronic progressive experimental autoimmune encephalomyelitis in nonobese diabetic mice, an experimental model that resembles several aspects of SPMS, including neurodegeneration and disease progression driven by the innate immune response in the CNS. Indeed, S1PR modulation by FTY720 in murine and human astrocytes suppressed neurodegeneration-promoting mechanisms mediated by astrocytes, microglia, and CNS-infiltrating proinflammatory monocytes. Genome-wide studies showed that FTY720 suppresses transcriptional programs associated with the promotion of disease progression by astrocytes. The study of the molecular mechanisms controlling these transcriptional modules may open new avenues for the development of therapeutic strategies for progressive MS.


Asunto(s)
Astrocitos/efectos de los fármacos , Inmunosupresores/farmacología , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Receptores de Lisoesfingolípidos/metabolismo , Animales , Astrocitos/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Femenino , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Inmunosupresores/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Microglía/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Esclerosis Múltiple Crónica Progresiva/patología , Cultivo Primario de Células , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Transcriptoma/efectos de los fármacos
5.
Trends Immunol ; 37(7): 427-439, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27236363

RESUMEN

The ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1, or CD39) catalyzes the phosphohydrolysis of extracellular ATP (eATP) and ADP (eADP) released under conditions of inflammatory stress and cell injury. CD39 generates AMP, which is in turn used by the ecto-5'-nucleotidase CD73 to synthesize adenosine. These ectonucleotidases have a major impact on the dynamic equilibrium of proinflammatory eATP and ADP nucleotides versus immunosuppressive adenosine nucleosides. Indeed, CD39 plays a dominant role in the purinergic regulation of inflammation and the immune response because its expression is influenced by genetic and environmental factors. We review the specific role of CD39 in the kinetic regulation of cellular immune responses in the evolution of disease. We focus on the effects of CD39 on T cells and explore potential clinical applications in autoimmunity, chronic infections, and cancer.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Enfermedades Autoinmunes/inmunología , Tolerancia Inmunológica/inmunología , Infecciones/inmunología , Inflamación/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , 5'-Nucleotidasa/metabolismo , Adenosina/metabolismo , Adenosina Monofosfato/metabolismo , Animales , Autoinmunidad , Interacción Gen-Ambiente , Humanos , Activación de Linfocitos
6.
J Immunol ; 196(2): 637-44, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26663782

RESUMEN

Despite accumulating evidence indicating that neurotransmitters released by the sympathetic nervous system can modulate the activity of innate immune cells, we still know very little about how norepinephrine impacts signaling pathways in dendritic cells (DC) and the consequence of that in DC-driven T cell differentiation. In this article, we demonstrate that ß2-adrenergic receptor (ß2AR) activation in LPS-stimulated DC does not impair their ability to promote T cell proliferation; however, it diminishes IL-12p70 secretion, leading to a shift in the IL-12p70/IL-23 ratio. Although ß2AR stimulation in DC induces protein kinase A-dependent cAMP-responsive element-binding protein phosphorylation, the effect of changing the profile of cytokines produced upon LPS challenge occurs in a protein kinase A-independent manner and, rather, is associated with inhibition of the NF-κB and AP-1 signaling pathways. Moreover, as a consequence of the inverted IL-12p70/IL-23 ratio following ß2AR stimulation, LPS-stimulated DC promoted the generation of CD4(+) T cells that, upon TCR engagement, produced lower amounts of IFN-γ and higher levels of IL-17. These findings provide new insights into molecular and cellular mechanisms by which ß2AR stimulation in murine DC can influence the generation of adaptive immune responses and may explain some aspects of how sympathetic nervous system activity can modulate immune function.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Norepinefrina/inmunología , Receptores Adrenérgicos beta 2/inmunología , Transducción de Señal/inmunología , Animales , Western Blotting , Diferenciación Celular/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción AP-1/inmunología
7.
J Immunol ; 190(6): 2614-21, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23390295

RESUMEN

We have previously shown that regulatory T (Treg) cells that accumulate in the airways of allergic mice upregulate CC-chemokine receptor 4 (CCR4) expression. These Treg cells suppressed in vitro Th2 cell proliferation but not type 2 cytokine production. In the current study, using a well-established murine model of allergic lung disease or oral tolerance, we evaluated the in vivo activity of Treg cells in allergic airway inflammation with special focus on CCR4 function. We found that allergic, but not tolerant, mice treated with anti-CD25 Ab showed increased airway eosinophilia and IL-5- or IL-4-producing Th2 cells when compared with untreated mice. Notably, mice with CCR4 deficiency displayed an augmented airway allergic inflammation compared with wild-type or CCR2 knockout (KO) mice. The allergic phenotype of CCR4KO mice was similar to that observed in anti-CD25-treated mice. The exacerbated allergic inflammation of CCR4KO mice was directly associated with an impaired migration of Treg cells to airways and augmented frequency of pulmonary Th2 cells. Adoptive transfer of CD25(+)CD4(+) T cells expressing high levels of CCR4, but not CCR4KO CD25(+)CD4(+) T cells, attenuated the severe airway Th2 response of CCR4KO mice. Our results show that CCR4 is critically involved in the migration of Treg cells to allergic lungs that, in turn, attenuate airway Th2 activation and allergic eosinophilic inflammation.


Asunto(s)
Movimiento Celular/inmunología , Eosinofilia/inmunología , Neumonía/inmunología , Receptores CCR4/fisiología , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/inmunología , Animales , Eosinofilia/genética , Eosinofilia/patología , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/genética , Neumonía/patología , Receptores CCR4/deficiencia , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Células Th2/inmunología , Células Th2/metabolismo , Células Th2/patología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
8.
Eur J Immunol ; 43(4): 1001-12, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23436577

RESUMEN

Beta2-adrenergic receptor (B2AR) signaling is known to impair Th1-cell differentiation and function in a cAMP-dependent way, leading to inhibition of cell proliferation and decreased production of IL-2 and IFN-γ. CD4(+) Foxp3(+) Treg cells play a key role in the regulation of immune responses and are essential for maintenance of self-tolerance. Nevertheless, very little is known about adrenergic receptor expression in Treg cells or the influence of noradrenaline on their function. Here we show that Foxp3(+) Treg cells express functional B2AR. B2AR activation in Treg cells leads to increased intracellular cAMP levels and to protein kinase A (PKA)-dependent CREB phosphorylation. We also found that signaling via B2AR enhances the in vitro suppressive activity of Treg cells. B2AR-mediated increase in Treg-cell suppressive function was associated with decreased IL-2 mRNA levels in responder CD4(+) T cells and improved Treg-cell-induced conversion of CD4(+) Foxp3(-) cells into Foxp3(+) induced Treg cells. Moreover, B2AR signaling increased CTLA-4 expression in Treg cells in a PKA-dependent way. Finally, we found that PKA inhibition totally prevented the B2AR-mediated increase in Treg-cell suppressive function. Our data suggest that sympathetic fibers are able to regulate Treg-cell suppressive activity in a positive manner through B2AR signaling.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Antígenos CD4/metabolismo , Antígeno CTLA-4/inmunología , Antígeno CTLA-4/metabolismo , Factores de Transcripción Forkhead/metabolismo , Interleucina-2/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados
9.
Front Immunol ; 14: 1267641, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38283348

RESUMEN

Introduction: Chagas disease causes a cardiac illness characterized by immunoinflammatory reactions leading to myocardial fibrosis and remodeling. The development of Chronic Chagas Cardiomyopathy (CCC) in some patients while others remain asymptomatic is not fully understood, but dysregulated inflammatory responses are implicated. The Aryl hydrocarbon receptor (AhR) plays a crucial role in regulating inflammation. Certain tryptophan (Trp) metabolites have been identified as AhR ligands with regulatory functions. Methods results and discussion: We investigated AhR expression, agonist response, ligand production, and AhR-dependent responses, such as IDO activation and regulatory T (Treg) cells induction, in two T. cruzi-infected mouse strains (B6 and Balb/c) showing different polymorphisms in AhR. Furthermore, we assessed the metabolic profile of Trp catabolites and AhR agonistic activity levels in plasma samples from patients with chronic Chagas disease (CCD) and healthy donors (HD) using a luciferase reporter assay and liquid chromatography-mass spectrophotometry (LC-MS) analysis. T. cruzi-infected B6 mice showed impaired AhR-dependent responses compared to Balb/c mice, including reduced IDO activity, kynurenine levels, Treg cell induction, CYP1A1 up-regulation, and AhR expression following agonist activation. Additionally, B6 mice exhibited no detectable AhR agonist activity in plasma and displayed lower CYP1A1 up-regulation and AhR expression upon agonist activation. Similarly, CCC patients had decreased AhR agonistic activity in plasma compared to HD patients and exhibited dysregulation in Trp metabolic pathways, resulting in altered plasma metabolite profiles. Notably, patients with severe CCC specifically showed increased N-acetylserotonin levels in their plasma. The methods and findings presented here contribute to a better understanding of CCC development mechanisms and may identify potential specific biomarkers for T. cruzi infection and the severity of associated heart disease. These insights could be valuable in designing new therapeutic strategies. Ultimately, this research aims to establish the AhR agonistic activity and Trp metabolic profile in plasma as an innovative, non-invasive predictor of prognosis for chronic Chagas disease.


Asunto(s)
Cardiomiopatía Chagásica , Enfermedad de Chagas , Animales , Humanos , Ratones , Enfermedad de Chagas/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Receptores de Hidrocarburo de Aril/agonistas , Triptófano/metabolismo
10.
Clin Immunol ; 142(2): 117-26, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22019771

RESUMEN

Some organ-transplanted patients achieve a state of "operational tolerance" (OT) in which graft function is maintained after the complete withdrawal of immunosuppressive drugs. We used a gene panel of regulatory/inflammatory molecules (FOXP3, GATA3, IL10, TGFB1, TGFBR1/ TBX21, TNF and IFNG) to investigate the gene expression profile in peripheral blood mononuclear cells of renal-transplanted individuals experiencing OT compared to transplanted individuals not displaying OT and healthy individuals (HI). OT subjects showed a predominant regulatory (REG) profile with higher gene expression of GATA3, FOXP3, TGFB1 and TGFB receptor 1 compared to the other groups. This predominant REG gene expression profile displayed stability over time. The significant GATA3 gene and protein expressions in OT individuals suggest that a Th2 deviation may be a relevant pathway to OT. Moreover, the capacity of the REG/INFLAMMA gene panel to discriminate OT by peripheral blood analysis indicates that this state has systemic repercussions.


Asunto(s)
Factor de Transcripción GATA3 , Inmunosupresores/metabolismo , Trasplante de Riñón/inmunología , Leucocitos Mononucleares/fisiología , Tolerancia al Trasplante , Adulto , Anciano , Femenino , Factores de Transcripción Forkhead/sangre , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción GATA3/sangre , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Perfilación de la Expresión Génica , Supervivencia de Injerto/inmunología , Humanos , Inmunosupresores/sangre , Masculino , Persona de Mediana Edad , Receptores de Factores de Crecimiento Transformadores beta/sangre , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Estudios Retrospectivos , Células Th2/metabolismo , Factor de Crecimiento Transformador beta1/sangre , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Tolerancia al Trasplante/genética , Tolerancia al Trasplante/inmunología
11.
Mol Med ; 18: 733-43, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22252714

RESUMEN

Transplanted individuals in operational tolerance (OT) maintain long-term stable graft function after completely stopping immunosuppression. Understanding the mechanisms involved in OT can provide valuable information about pathways to human transplantation tolerance. Here we report that operationally tolerant individuals display quantitative and functional preservation of the B-cell compartment in renal transplantation. OT exhibited normal numbers of circulating total B cells, naive, memory and regulatory B cells (Bregs) as well as preserved B-cell receptor repertoire, similar to healthy individuals. In addition, OT also displayed conserved capacity to activate the cluster of differentiation 40 (CD40)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in Bregs, in contrast, with chronic rejection. Rather than expansion or higher activation, we show that the preservation of the B-cell compartment favors OT.


Asunto(s)
Linfocitos B/inmunología , Trasplante de Riñón/inmunología , Tolerancia al Trasplante/inmunología , Adulto , Anciano , Linfocitos B/metabolismo , Linfocitos B Reguladores/inmunología , Linfocitos B Reguladores/metabolismo , Antígenos CD40/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
12.
Clin Dev Immunol ; 2012: 721817, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22162718

RESUMEN

Foxp3(+)CD25(+)CD4(+) regulatory T cells are vital for peripheral tolerance and control of tissue inflammation. In this study, we characterized the phenotype and monitored the migration and activity of regulatory T cells present in the airways of allergic or tolerant mice after allergen challenge. To induce lung allergic inflammation, mice were sensitized twice with ovalbumin/aluminum hydroxide gel and challenged twice with intranasal ovalbumin. Tolerance was induced by oral administration of ovalbumin for 5 consecutive days prior to OVA sensitization and challenge. We detected regulatory T cells (Foxp3(+)CD25(+)CD4(+) T cells) in the airways of allergic and tolerant mice; however, the number of regulatory T cells was more than 40-fold higher in allergic mice than in tolerant mice. Lung regulatory T cells expressed an effector/memory phenotype (CCR4(high)CD62L(low)CD44(high)CD54(high)CD69(+)) that distinguished them from naive regulatory T cells (CCR4(int)CD62L(high)CD44(int)CD54(int)CD69(-)). These regulatory T cells efficiently suppressed pulmonary T-cell proliferation but not Th2 cytokine production.


Asunto(s)
Asma/inmunología , Proliferación Celular , Citocinas/biosíntesis , Pulmón/inmunología , Neumonía/inmunología , Linfocitos T Reguladores/inmunología , Animales , Asma/patología , Antígenos CD4/metabolismo , Femenino , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Neumonía/patología , Células Th2/inmunología , Células Th2/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-33408169

RESUMEN

OBJECTIVE: MS is an autoimmune demyelinating disease of the CNS, which causes neurologic deficits in young adults and leads to progressive disability. The aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor, can drive anti-inflammatory functions in peripheral immune cells and also in CNS-resident cells. Laquinimod is a drug developed for the treatment of MS known to activate AHR, but the cellular targets of laquinimod are still not completely known. In this work, we analyzed the contribution of AHR activation in astrocytes to its beneficial effects in the experimental autoimmune encephalomyelitis (EAE) preclinical model of MS. METHODS: We used conditional knockout mice, in combination with genome-wide analysis of gene expression by RNA-seq and in vitro culture systems to investigate the effects of laquinimod on astrocytes. RESULTS: We found that AHR activation in astrocytes by laquinimod ameliorates EAE, a preclinical model of MS. Genome-wide RNA-seq transcriptional analyses detected anti-inflammatory effects of laquinimod in glial cells during EAE. Moreover, we established that the Delaq metabolite of laquinimod dampens proinflammatory mediator production while activating tissue-protective mechanisms in glia. CONCLUSIONS: Taken together, these findings suggest that AHR activation by clinically relevant AHR agonists may represent a novel therapeutic approach for the treatment of MS.


Asunto(s)
Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Quinolonas/uso terapéutico , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinolonas/farmacología , Receptores de Hidrocarburo de Aril/inmunología
15.
Nat Neurosci ; 23(8): 939-951, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32690969

RESUMEN

Zika virus (ZIKV) is a flavivirus linked to multiple birth defects including microcephaly, known as congenital ZIKV syndrome. The identification of host factors involved in ZIKV replication may guide efficacious therapeutic interventions. In genome-wide transcriptional studies, we found that ZIKV infection triggers aryl hydrocarbon receptor (AHR) activation. Specifically, ZIKV infection induces kynurenine (Kyn) production, which activates AHR, limiting the production of type I interferons (IFN-I) involved in antiviral immunity. Moreover, ZIKV-triggered AHR activation suppresses intrinsic immunity driven by the promyelocytic leukemia (PML) protein, which limits ZIKV replication. AHR inhibition suppressed the replication of multiple ZIKV strains in vitro and also suppressed replication of the related flavivirus dengue. Finally, AHR inhibition with a nanoparticle-delivered AHR antagonist or an inhibitor developed for human use limited ZIKV replication and ameliorated newborn microcephaly in a murine model. In summary, we identified AHR as a host factor for ZIKV replication and PML protein as a driver of anti-ZIKV intrinsic immunity.


Asunto(s)
Receptores de Hidrocarburo de Aril/metabolismo , Replicación Viral , Virus Zika/metabolismo , Animales , Chlorocebus aethiops , Células Hep G2 , Humanos , Células Vero , Infección por el Virus Zika/metabolismo
16.
Cell Rep ; 28(12): 3120-3130.e5, 2019 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-31533035

RESUMEN

Noradrenaline (NE), the main neurotransmitter released by sympathetic nerve terminals, is known to modulate the immune response. However, the role of the sympathetic nervous system (SNS) on the development of autoimmune diseases is still unclear. Here, we report that the SNS limits the generation of pathogenic T cells and disease development in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS). ß2-Adrenergic receptor (Adrb2) signaling limits T cell autoimmunity in EAE through a mechanism mediated by the suppression of IL-2, IFN-γ, and GM-CSF production via inducible cAMP early repressor (ICER). Accordingly, the lack of Adrb2 signaling in immune cells is sufficient to abrogate the suppressive effects of SNS activity, resulting in increased pathogenic T cell responses and EAE development. Collectively, these results uncover a suppressive role for the SNS in CNS autoimmunity while they identify potential targets for therapeutic intervention.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Inmunidad Celular , Esclerosis Múltiple/inmunología , Receptores Adrenérgicos beta 2/inmunología , Transducción de Señal/inmunología , Sistema Nervioso Simpático/inmunología , Linfocitos T/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Receptores Adrenérgicos beta 2/genética , Transducción de Señal/genética , Sistema Nervioso Simpático/patología , Linfocitos T/patología
17.
Nat Neurosci ; 22(5): 729-740, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962630

RESUMEN

Tumor-associated macrophages (TAMs) play an important role in the immune response to cancer, but the mechanisms by which the tumor microenvironment controls TAMs and T cell immunity are not completely understood. Here we report that kynurenine produced by glioblastoma cells activates aryl hydrocarbon receptor (AHR) in TAMs to modulate their function and T cell immunity. AHR promotes CCR2 expression, driving TAM recruitment in response to CCL2. AHR also drives the expression of KLF4 and suppresses NF-κB activation in TAMs. Finally, AHR drives the expression of the ectonucleotidase CD39 in TAMs, which promotes CD8+ T cell dysfunction by producing adenosine in cooperation with CD73. In humans, the expression of AHR and CD39 was highest in grade 4 glioma, and high AHR expression was associated with poor prognosis. In summary, AHR and CD39 expressed in TAMs participate in the regulation of the immune response in glioblastoma and constitute potential targets for immunotherapy.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Quinurenina/metabolismo , Macrófagos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Glioblastoma/metabolismo , Humanos , Factor 4 Similar a Kruppel , Receptores de Lipopolisacáridos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Factor de Transcripción STAT1 , Factor de Transcripción STAT3/metabolismo , Linfocitos T/inmunología , Microambiente Tumoral
19.
Semin Immunopathol ; 39(2): 113-120, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27646959

RESUMEN

Deficits in immunological tolerance against self-antigens and antigens provided by the diet and commensal microbiota can result in the development of inflammatory and autoimmune disorders. Dendritic cells (DCs) are pivotal regulators of the immune response, specialized in antigen presentation to drive T cell priming and differentiation. DCs also have a tolerogenic function, participating in the enforcement of central and peripheral tolerance and the resolution of ongoing immune responses. Thus, DCs control effector and regulatory mechanisms relevant to the pathology of autoimmune disorders. In this review, we discuss recent findings regarding the control of the adaptive immune response by tolerogenic DCs. A thorough understanding of the mechanisms that control the tolerogenic DC phenotype will guide the development of novel strategies for the treatment of autoimmunity.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica , Animales , Presentación de Antígeno/inmunología , Autoinmunidad , Biomarcadores , Diferenciación Celular , Linaje de la Célula , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Expresión Génica , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Perforina/genética , Receptores Colinérgicos/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
20.
Semin Immunopathol ; 39(2): 165-176, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27800584

RESUMEN

Dendritic cells are of paramount importance bridging innate and adaptive immune responses. Depending on the context, after sensing environmental antigens, commensal microorganisms, pathogenic agents, or antigens from the diet, dendritic cells may drive either different effector adaptive immune responses or tolerance, avoiding tissue damage. Although the plasticity of the immune response and the capacity to regulate itself are considered essential to orchestrate appropriate physiological responses, it is known that the nervous system plays a relevant role controlling immune cell function. Dendritic cells present in the skin, the intestine, and lymphoid organs, besides expressing adrenergic receptors, can be reached by neurotransmitters released by sympathetic fibers innervating these tissues. These review focus on how neurotransmitters from the sympathetic nervous system can modulate dendritic cell function and how this may impact the immune response and immune-mediated disorders.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Sistema Inmunológico/citología , Sistema Inmunológico/fisiología , Neuroinmunomodulación , Sistema Nervioso Simpático/fisiología , Animales , Citocinas/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/inervación , Tejido Linfoide/inmunología , Tejido Linfoide/inervación , Tejido Linfoide/metabolismo , Norepinefrina/metabolismo , Receptores Adrenérgicos/metabolismo , Transducción de Señal , Piel/inmunología , Piel/inervación , Piel/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA