Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 275
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 39: 251-277, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33556248

RESUMEN

The immune system of the central nervous system (CNS) consists primarily of innate immune cells. These are highly specialized macrophages found either in the parenchyma, called microglia, or at the CNS interfaces, such as leptomeningeal, perivascular, and choroid plexus macrophages. While they were primarily thought of as phagocytes, their function extends well beyond simple removal of cell debris during development and diseases. Brain-resident innate immune cells were found to be plastic, long-lived, and host to an outstanding number of risk genes for multiple pathologies. As a result, they are now considered the most suitable targets for modulating CNS diseases. Additionally, recent single-cell technologies enhanced our molecular understanding of their origins, fates, interactomes, and functional cell statesduring health and perturbation. Here, we review the current state of our understanding and challenges of the myeloid cell biology in the CNS and treatment options for related diseases.


Asunto(s)
Sistema Nervioso Central , Microglía , Animales , Encéfalo , Humanos , Macrófagos , Células Mieloides
2.
Cell ; 186(7): 1309-1327, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-37001498

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of the central nervous system afflicting nearly three million individuals worldwide. Neuroimmune interactions between glial, neural, and immune cells play important roles in MS pathology and offer potential targets for therapeutic intervention. Here, we review underlying risk factors, mechanisms of MS pathogenesis, available disease modifying therapies, and examine the value of emerging technologies, which may address unmet clinical needs and identify novel therapeutic targets.


Asunto(s)
Esclerosis Múltiple , Humanos , Esclerosis Múltiple/tratamiento farmacológico , Sistema Nervioso Central , Neuroglía , Fenómenos Fisiológicos Celulares , Inflamación/patología
3.
Nat Immunol ; 25(3): 432-447, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38409259

RESUMEN

Central nervous system (CNS)-resident cells such as microglia, oligodendrocytes and astrocytes are gaining increasing attention in respect to their contribution to CNS pathologies including multiple sclerosis (MS). Several studies have demonstrated the involvement of pro-inflammatory glial subsets in the pathogenesis and propagation of inflammatory events in MS and its animal models. However, it has only recently become clear that the underlying heterogeneity of astrocytes and microglia can not only drive inflammation, but also lead to its resolution through direct and indirect mechanisms. Failure of these tissue-protective mechanisms may potentiate disease and increase the risk of conversion to progressive stages of MS, for which currently available therapies are limited. Using proteomic analyses of cerebrospinal fluid specimens from patients with MS in combination with experimental studies, we here identify Heparin-binding EGF-like growth factor (HB-EGF) as a central mediator of tissue-protective and anti-inflammatory effects important for the recovery from acute inflammatory lesions in CNS autoimmunity. Hypoxic conditions drive the rapid upregulation of HB-EGF by astrocytes during early CNS inflammation, while pro-inflammatory conditions suppress trophic HB-EGF signaling through epigenetic modifications. Finally, we demonstrate both anti-inflammatory and tissue-protective effects of HB-EGF in a broad variety of cell types in vitro and use intranasal administration of HB-EGF in acute and post-acute stages of autoimmune neuroinflammation to attenuate disease in a preclinical mouse model of MS. Altogether, we identify astrocyte-derived HB-EGF and its epigenetic regulation as a modulator of autoimmune CNS inflammation and potential therapeutic target in MS.


Asunto(s)
Astrocitos , Esclerosis Múltiple , Animales , Humanos , Ratones , Antiinflamatorios , Modelos Animales de Enfermedad , Epigénesis Genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Inflamación , Proteómica
4.
Cell ; 176(3): 581-596.e18, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30661753

RESUMEN

Genome-wide studies have identified genetic variants linked to neurologic diseases. Environmental factors also play important roles, but no methods are available for their comprehensive investigation. We developed an approach that combines genomic data, screens in a novel zebrafish model, computational modeling, perturbation studies, and multiple sclerosis (MS) patient samples to evaluate the effects of environmental exposure on CNS inflammation. We found that the herbicide linuron amplifies astrocyte pro-inflammatory activities by activating signaling via sigma receptor 1, inositol-requiring enzyme-1α (IRE1α), and X-box binding protein 1 (XBP1). Indeed, astrocyte-specific shRNA- and CRISPR/Cas9-driven gene inactivation combined with RNA-seq, ATAC-seq, ChIP-seq, and study of patient samples suggest that IRE1α-XBP1 signaling promotes CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, MS. In summary, these studies define environmental mechanisms that control astrocyte pathogenic activities and establish a multidisciplinary approach for the systematic investigation of the effects of environmental exposure in neurologic disorders.


Asunto(s)
Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Animales , Sistema Nervioso Central/inmunología , Biología Computacional/métodos , Encefalomielitis Autoinmune Experimental/inmunología , Endorribonucleasas/metabolismo , Ambiente , Exposición a Riesgos Ambientales/efectos adversos , Genoma , Genómica , Humanos , Inflamación/metabolismo , Linurona/efectos adversos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores sigma/efectos de los fármacos , Receptores sigma/metabolismo , Transducción de Señal , Proteína 1 de Unión a la X-Box/metabolismo , Pez Cebra
5.
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
6.
Immunity ; 56(9): 1983-1985, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37703827

RESUMEN

Many mechanisms by which stress mediates its effects within the central nervous system still remain unknown. Byun, Kim, Kim et al. find that early-life stress triggers corticosterone release to drive astrocyte-dependent synapse elimination and altered behavior. Thus, this work defines a steroid-sensitive astrocyte transcriptional circuit controlling behavior, highlighting how the study of CNS immunoregulation may shed light on behavior.


Asunto(s)
Astrocitos , Sinapsis , Sistema Nervioso Central , Esteroides
7.
Nature ; 627(8005): 865-872, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38509377

RESUMEN

Disease-associated astrocyte subsets contribute to the pathology of neurologic diseases, including multiple sclerosis and experimental autoimmune encephalomyelitis1-8 (EAE), an experimental model for multiple sclerosis. However, little is known about the stability of these astrocyte subsets and their ability to integrate past stimulation events. Here we report the identification of an epigenetically controlled memory astrocyte subset that exhibits exacerbated pro-inflammatory responses upon rechallenge. Specifically, using a combination of single-cell RNA sequencing, assay for transposase-accessible chromatin with sequencing, chromatin immunoprecipitation with sequencing, focused interrogation of cells by nucleic acid detection and sequencing, and cell-specific in vivo CRISPR-Cas9-based genetic perturbation studies we established that astrocyte memory is controlled by the metabolic enzyme ATP-citrate lyase (ACLY), which produces acetyl coenzyme A (acetyl-CoA) that is used by histone acetyltransferase p300 to control chromatin accessibility. The number of ACLY+p300+ memory astrocytes is increased in acute and chronic EAE models, and their genetic inactivation ameliorated EAE. We also detected the pro-inflammatory memory phenotype in human astrocytes in vitro; single-cell RNA sequencing and immunohistochemistry studies detected increased numbers of ACLY+p300+ astrocytes in chronic multiple sclerosis lesions. In summary, these studies define an epigenetically controlled memory astrocyte subset that promotes CNS pathology in EAE and, potentially, multiple sclerosis. These findings may guide novel therapeutic approaches for multiple sclerosis and other neurologic diseases.


Asunto(s)
Astrocitos , Encefalomielitis Autoinmune Experimental , Memoria Epigenética , Esclerosis Múltiple , Animales , Femenino , Humanos , Masculino , Ratones , Acetilcoenzima A/metabolismo , Astrocitos/enzimología , Astrocitos/metabolismo , Astrocitos/patología , ATP Citrato (pro-S)-Liasa/metabolismo , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Sistemas CRISPR-Cas , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Inflamación/enzimología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Esclerosis Múltiple/enzimología , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Análisis de Expresión Génica de una Sola Célula , Transposasas/metabolismo
8.
Nature ; 614(7947): 318-325, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36599978

RESUMEN

Rare CD4 T cells that contain HIV under antiretroviral therapy represent an important barrier to HIV cure1-3, but the infeasibility of isolating and characterizing these cells in their natural state has led to uncertainty about whether they possess distinctive attributes that HIV cure-directed therapies might exploit. Here we address this challenge using a microfluidic technology that isolates the transcriptomes of HIV-infected cells based solely on the detection of HIV DNA. HIV-DNA+ memory CD4 T cells in the blood from people receiving antiretroviral therapy showed inhibition of six transcriptomic pathways, including death receptor signalling, necroptosis signalling and antiproliferative Gα12/13 signalling. Moreover, two groups of genes identified by network co-expression analysis were significantly associated with HIV-DNA+ cells. These genes (n = 145) accounted for just 0.81% of the measured transcriptome and included negative regulators of HIV transcription that were higher in HIV-DNA+ cells, positive regulators of HIV transcription that were lower in HIV-DNA+ cells, and other genes involved in RNA processing, negative regulation of mRNA translation, and regulation of cell state and fate. These findings reveal that HIV-infected memory CD4 T cells under antiretroviral therapy are a distinctive population with host gene expression patterns that favour HIV silencing, cell survival and cell proliferation, with important implications for the development of HIV cure strategies.


Asunto(s)
Linfocitos T CD4-Positivos , Regulación Viral de la Expresión Génica , Infecciones por VIH , VIH-1 , Latencia del Virus , Humanos , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , ADN Viral/aislamiento & purificación , Regulación Viral de la Expresión Génica/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/genética , VIH-1/aislamiento & purificación , VIH-1/patogenicidad , Memoria Inmunológica , Microfluídica , Necroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Antirretrovirales/farmacología , Antirretrovirales/uso terapéutico
9.
Nature ; 614(7947): 326-333, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36599367

RESUMEN

Multiple sclerosis is a chronic inflammatory disease of the central nervous system1. Astrocytes are heterogeneous glial cells that are resident in the central nervous system and participate in the pathogenesis of multiple sclerosis and its model experimental autoimmune encephalomyelitis2,3. However, few unique surface markers are available for the isolation of astrocyte subsets, preventing their analysis and the identification of candidate therapeutic targets; these limitations are further amplified by the rarity of pathogenic astrocytes. Here, to address these challenges, we developed focused interrogation of cells by nucleic acid detection and sequencing (FIND-seq), a high-throughput microfluidic cytometry method that combines encapsulation of cells in droplets, PCR-based detection of target nucleic acids and droplet sorting to enable in-depth transcriptomic analyses of cells of interest at single-cell resolution. We applied FIND-seq to study the regulation of astrocytes characterized by the splicing-driven activation of the transcription factor XBP1, which promotes disease pathology in multiple sclerosis and experimental autoimmune encephalomyelitis4. Using FIND-seq in combination with conditional-knockout mice, in vivo CRISPR-Cas9-driven genetic perturbation studies and bulk and single-cell RNA sequencing analyses of samples from mouse experimental autoimmune encephalomyelitis and humans with multiple sclerosis, we identified a new role for the nuclear receptor NR3C2 and its corepressor NCOR2 in limiting XBP1-driven pathogenic astrocyte responses. In summary, we used FIND-seq to identify a therapeutically targetable mechanism that limits XBP1-driven pathogenic astrocyte responses. FIND-seq enables the investigation of previously inaccessible cells, including rare cell subsets defined by unique gene expression signatures or other nucleic acid markers.


Asunto(s)
Astrocitos , Encefalomielitis Autoinmune Experimental , Microfluídica , Esclerosis Múltiple , Ácidos Nucleicos , Análisis de Expresión Génica de una Sola Célula , Animales , Humanos , Ratones , Astrocitos/metabolismo , Astrocitos/patología , Regulación de la Expresión Génica , Ratones Noqueados , Esclerosis Múltiple/patología , Microfluídica/métodos , Análisis de Expresión Génica de una Sola Célula/métodos , Ácidos Nucleicos/análisis , Edición Génica
10.
Nature ; 620(7975): 881-889, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37558878

RESUMEN

Dendritic cells (DCs) have a role in the development and activation of self-reactive pathogenic T cells1,2. Genetic variants that are associated with the function of DCs have been linked to autoimmune disorders3,4, and DCs are therefore attractive therapeutic targets for such diseases. However, developing DC-targeted therapies for autoimmunity requires identification of the mechanisms that regulate DC function. Here, using single-cell and bulk transcriptional and metabolic analyses in combination with cell-specific gene perturbation studies, we identify a regulatory loop of negative feedback that operates in DCs to limit immunopathology. Specifically, we find that lactate, produced by activated DCs and other immune cells, boosts the expression of NDUFA4L2 through a mechanism mediated by hypoxia-inducible factor 1α (HIF-1α). NDUFA4L2 limits the production of mitochondrial reactive oxygen species that activate XBP1-driven transcriptional modules in DCs that are involved in the control of pathogenic autoimmune T cells. We also engineer a probiotic that produces lactate and suppresses T cell autoimmunity through the activation of HIF-1α-NDUFA4L2 signalling in DCs. In summary, we identify an immunometabolic pathway that regulates DC function, and develop a synthetic probiotic for its therapeutic activation.


Asunto(s)
Enfermedades Autoinmunes , Sistema Nervioso Central , Células Dendríticas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ácido Láctico , Humanos , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/prevención & control , Autoinmunidad , Sistema Nervioso Central/citología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ácido Láctico/metabolismo , Probióticos/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/inmunología , Retroalimentación Fisiológica , Lactasa/genética , Lactasa/metabolismo , Análisis de la Célula Individual
11.
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
12.
Nature ; 590(7846): 473-479, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33408417

RESUMEN

Astrocytes are glial cells that are abundant in the central nervous system (CNS) and that have important homeostatic and disease-promoting functions1. However, little is known about the homeostatic anti-inflammatory activities of astrocytes and their regulation. Here, using high-throughput flow cytometry screening, single-cell RNA sequencing and CRISPR-Cas9-based cell-specific in vivo genetic perturbations in mice, we identify a subset of astrocytes that expresses the lysosomal protein LAMP12 and the death receptor ligand TRAIL3. LAMP1+TRAIL+ astrocytes limit inflammation in the CNS by inducing T cell apoptosis through TRAIL-DR5 signalling. In homeostatic conditions, the expression of TRAIL in astrocytes is driven by interferon-γ (IFNγ) produced by meningeal natural killer (NK) cells, in which IFNγ expression is modulated by the gut microbiome. TRAIL expression in astrocytes is repressed by molecules produced by T cells and microglia in the context of inflammation. Altogether, we show that LAMP1+TRAIL+ astrocytes limit CNS inflammation by inducing T cell apoptosis, and that this astrocyte subset is maintained by meningeal IFNγ+ NK cells that are licensed by the microbiome.


Asunto(s)
Astrocitos/inmunología , Microbioma Gastrointestinal/inmunología , Inflamación/prevención & control , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Proteínas de Membrana de los Lisosomas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Apoptosis , Astrocitos/metabolismo , Biomarcadores , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Homeostasis , Humanos , Inflamación/inmunología , Meninges/citología , Meninges/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/citología , Linfocitos T/inmunología
13.
Cell ; 145(7): 1142-55, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21664664

RESUMEN

Neurogenesis and gliogenesis continue in discrete regions of the adult mammalian brain. A fundamental question remains whether cell genesis occurs from distinct lineage-restricted progenitors or from self-renewing and multipotent neural stem cells in the adult brain. Here, we developed a genetic marking strategy for lineage tracing of individual, quiescent, and nestin-expressing radial glia-like (RGL) precursors in the adult mouse dentate gyrus. Clonal analysis identified multiple modes of RGL activation, including asymmetric and symmetric self-renewal. Long-term lineage tracing in vivo revealed a significant percentage of clones that contained RGL(s), neurons, and astrocytes, indicating capacity of individual RGLs for both self-renewal and multilineage differentiation. Furthermore, conditional Pten deletion in RGLs initially promotes their activation and symmetric self-renewal but ultimately leads to terminal astrocytic differentiation and RGL depletion in the adult hippocampus. Our study identifies RGLs as self-renewing and multipotent neural stem cells and provides novel insights into in vivo properties of adult neural stem cells.


Asunto(s)
Células Madre Adultas/citología , Hipocampo/citología , Células Madre Multipotentes/citología , Células-Madre Neurales/citología , Neurogénesis , Animales , Giro Dentado/citología , Proteínas de Filamentos Intermediarios/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Nestina
14.
Nature ; 578(7796): 593-599, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051591

RESUMEN

Multiple sclerosis is a chronic inflammatory disease of the CNS1. Astrocytes contribute to the pathogenesis of multiple sclerosis2, but little is known about the heterogeneity of astrocytes and its regulation. Here we report the analysis of astrocytes in multiple sclerosis and its preclinical model experimental autoimmune encephalomyelitis (EAE) by single-cell RNA sequencing in combination with cell-specific Ribotag RNA profiling, assay for transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunoprecipitation with sequencing (ChIP-seq), genome-wide analysis of DNA methylation and in vivo CRISPR-Cas9-based genetic perturbations. We identified astrocytes in EAE and multiple sclerosis that were characterized by decreased expression of NRF2 and increased expression of MAFG, which cooperates with MAT2α to promote DNA methylation and represses antioxidant and anti-inflammatory transcriptional programs. Granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling in astrocytes drives the expression of MAFG and MAT2α and pro-inflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, multiple sclerosis. Our results identify candidate therapeutic targets in multiple sclerosis.


Asunto(s)
Astrocitos/patología , Sistema Nervioso Central/patología , Inflamación/patología , Factor de Transcripción MafG/genética , Proteínas Represoras/genética , Animales , Antioxidantes/metabolismo , Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Metilación de ADN , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inflamación/genética , Masculino , Metionina Adenosiltransferasa/genética , Ratones , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Factor 2 Relacionado con NF-E2/genética , Análisis de Secuencia de ARN , Transducción de Señal , Transcripción Genética
15.
Nature ; 586(7829): 417-423, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32999463

RESUMEN

Microglia, the brain's resident macrophages, help to regulate brain function by removing dying neurons, pruning non-functional synapses, and producing ligands that support neuronal survival1. Here we show that microglia are also critical modulators of neuronal activity and associated behavioural responses in mice. Microglia respond to neuronal activation by suppressing neuronal activity, and ablation of microglia amplifies and synchronizes the activity of neurons, leading to seizures. Suppression of neuronal activation by microglia occurs in a highly region-specific fashion and depends on the ability of microglia to sense and catabolize extracellular ATP, which is released upon neuronal activation by neurons and astrocytes. ATP triggers the recruitment of microglial protrusions and is converted by the microglial ATP/ADP hydrolysing ectoenzyme CD39 into AMP; AMP is then converted into adenosine by CD73, which is expressed on microglia as well as other brain cells. Microglial sensing of ATP, the ensuing microglia-dependent production of adenosine, and the adenosine-mediated suppression of neuronal responses via the adenosine receptor A1R are essential for the regulation of neuronal activity and animal behaviour. Our findings suggest that this microglia-driven negative feedback mechanism operates similarly to inhibitory neurons and is essential for protecting the brain from excessive activation in health and disease.


Asunto(s)
Retroalimentación Fisiológica , Microglía/fisiología , Inhibición Neural , Neuronas/fisiología , 5'-Nucleotidasa/metabolismo , Potenciales de Acción , Adenosina/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antígenos CD/metabolismo , Apirasa/metabolismo , Calcio/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Inhibición Neural/genética , Receptor de Adenosina A1/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Factores de Tiempo
16.
Nature ; 627(8003): 277-279, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38448528
18.
Bioinformatics ; 39(9)2023 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-37610350

RESUMEN

MOTIVATION: The method of genome-wide association studies (GWAS) and metabolomics combined provide an quantitative approach to pinpoint metabolic pathways and genes linked to specific diseases; however, such analyses require both genomics and metabolomics datasets from the same individuals/samples. In most cases, this approach is not feasible due to high costs, lack of technical infrastructure, unavailability of samples, and other factors. Therefore, an unmet need exists for a bioinformatics tool that can identify gene loci-associated polymorphic variants for metabolite alterations seen in disease states using standalone metabolomics. RESULTS: Here, we developed a bioinformatics tool, metGWAS 1.0, that integrates independent GWAS data from the GWAS database and standalone metabolomics data using a network-based systems biology approach to identify novel disease/trait-specific metabolite-gene associations. The tool was evaluated using standalone metabolomics datasets extracted from two metabolomics-GWAS case studies. It discovered both the observed and novel gene loci with known single nucleotide polymorphisms when compared to the original studies. AVAILABILITY AND IMPLEMENTATION: The developed metGWAS 1.0 framework is implemented in an R pipeline and available at: https://github.com/saifurbd28/metGWAS-1.0.


Asunto(s)
Estudio de Asociación del Genoma Completo , Metabolómica , Humanos , Flujo de Trabajo , Biología Computacional , Bases de Datos Factuales
19.
Environ Sci Technol ; 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38832692

RESUMEN

Cold heavy oil production with sand (CHOPS) is an extraction process for heavy oil in Canada, with the potential to lead to higher CH4 venting than conventional oil sites, that have not been adequately characterized. In order to quantify CH4 emissions from CHOPS activities, a focused aerial measurement campaign was conducted in the Canadian provinces of Alberta and Saskatchewan in June 2018. Total CH4 emissions from each of 10 clusters of CHOPS wells (containing 22-167 well sites per cluster) were derived using a mass balance computation algorithm that uses in situ wind data measurement on board aircraft. Results show that there is no statistically significant difference in CH4 emissions from CHOPS wells between the two provinces. Cluster-aggregated emission factors (EF) were determined using correspondingly aggregated production volumes. The average CH4 EF was 70.4 ± 36.9 kg/m3 produced oil for the Alberta wells and 55.1 ± 13.7 kg/m3 produced oil for the Saskatchewan wells. Using these EF and heavy oil production volumes reported to provincial regulators, the annual CH4 emissions from CHOPS were estimated to be 121% larger than CHOPS emissions extracted from Canada's National Inventory Report (NIR) for Saskatchewan. The EF were found to be positively correlated with the percentage of nonpiped production volumes in each cluster, indicating higher emissions for nonpiped wells while suggesting an avenue for methane emission reductions. A comparison with recent measurements indicates relatively limited effectiveness of regulations for Saskatchewan compared to those in Alberta. The results of this study indicate the substantial contribution of CHOPS operations to the underreporting observed in the NIR and provide measurement-based EF that can be used to develop improved emissions inventories for this sector and mitigate CH4 emissions from CHOPS operations.

20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA