Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 12, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38178091

RESUMEN

The hypothalamus is a brain structure that is deputed to maintain organism homeostasis by regulating autonomic function and hormonal production as part of the neuroendocrine system. Dysfunction in hypothalamic activity results in behavioral alterations, depression, metabolic syndromes, fatigue, and infertility. Remarkably, many of these symptoms are associated with multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS) characterized by focal demyelination, immune cell infiltration into the brain parenchyma, and neurodegeneration. Furthermore, altered hormonal levels have been documented in MS patients, suggesting the putative involvement of hypothalamic deficits in MS clinical manifestations. Yet, a systematic analysis of hypothalamic function in response to neuroinflammatory stress is still lacking. To fill this gap, here we performed a longitudinal profiling of the hypothalamic transcriptome upon experimental autoimmune encephalomyelitis (EAE)-a murine disease model recapitulating key MS phenotypes at both histopathological and molecular levels. We show that changes in gene expression connected with an anti-inflammatory response start already at pre-onset and persist along EAE progression. Altered levels of hypothalamic neuropeptides were also detected, which possibly underlie homeostatic responses to stress and aberrant feeding behaviors. Last, a thorough investigation of the principal endocrine glands highlighted defects in the main steroidogenic pathways upon disease. Collectively, our findings corroborate the central role of hypothalamic dysfunction in CNS autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Humanos , Ratones , Animales , Transcriptoma , Encefalomielitis Autoinmune Experimental/patología , Esclerosis Múltiple/patología , Sistema Nervioso Central/patología , Hipotálamo/metabolismo , Ratones Endogámicos C57BL
2.
Brain ; 146(2): 645-656, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-35253861

RESUMEN

Polygenic inheritance plays a pivotal role in driving multiple sclerosis susceptibility, an inflammatory demyelinating disease of the CNS. We developed polygenic risk scores (PRS) of multiple sclerosis and assessed associations with both disease status and severity in cohorts of European descent. The largest genome-wide association dataset for multiple sclerosis to date (n = 41 505) was leveraged to generate PRS scores, serving as an informative susceptibility marker, tested in two independent datasets, UK Biobank [area under the curve (AUC) = 0.73, 95% confidence interval (CI): 0.72-0.74, P = 6.41 × 10-146] and Kaiser Permanente in Northern California (KPNC, AUC = 0.8, 95% CI: 0.76-0.82, P = 1.5 × 10-53). Individuals within the top 10% of PRS were at higher than 5-fold increased risk in UK Biobank (95% CI: 4.7-6, P = 2.8 × 10-45) and 15-fold higher risk in KPNC (95% CI: 10.4-24, P = 3.7 × 10-11), relative to the median decile. The cumulative absolute risk of developing multiple sclerosis from age 20 onwards was significantly higher in genetically predisposed individuals according to PRS. Furthermore, inclusion of PRS in clinical risk models increased the risk discrimination by 13% to 26% over models based only on conventional risk factors in UK Biobank and KPNC, respectively. Stratifying disease risk by gene sets representative of curated cellular signalling cascades, nominated promising genetic candidate programmes for functional characterization. These pathways include inflammatory signalling mediation, response to viral infection, oxidative damage, RNA polymerase transcription, and epigenetic regulation of gene expression to be among significant contributors to multiple sclerosis susceptibility. This study also indicates that PRS is a useful measure for estimating susceptibility within related individuals in multicase families. We show a significant association of genetic predisposition with thalamic atrophy within 10 years of disease progression in the UCSF-EPIC cohort (P < 0.001), consistent with a partial overlap between the genetics of susceptibility and end-organ tissue injury. Mendelian randomization analysis suggested an effect of multiple sclerosis susceptibility on thalamic volume, which was further indicated to be through horizontal pleiotropy rather than a causal effect. In summary, this study indicates important, replicable associations of PRS with enhanced risk assessment and radiographic outcomes of tissue injury, potentially informing targeted screening and prevention strategies.


Asunto(s)
Estudio de Asociación del Genoma Completo , Esclerosis Múltiple , Humanos , Herencia Multifactorial/genética , Esclerosis Múltiple/genética , Epigénesis Genética , Pueblo Europeo , Factores de Riesgo , Predisposición Genética a la Enfermedad/genética , Fenotipo
3.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34911760

RESUMEN

Epigenetic changes have been consistently detected in different cell types in multiple sclerosis (MS). However, their contribution to MS pathogenesis remains poorly understood partly because of sample heterogeneity and limited coverage of array-based methods. To fill this gap, we conducted a comprehensive analysis of genome-wide DNA methylation patterns in four peripheral immune cell populations isolated from 29 MS patients at clinical disease onset and 24 healthy controls. We show that B cells from new-onset untreated MS cases display more significant methylation changes than other disease-implicated immune cell types, consisting of a global DNA hypomethylation signature. Importantly, 4,933 MS-associated differentially methylated regions in B cells were identified, and this epigenetic signature underlies specific genetic programs involved in B cell differentiation and activation. Integration of the methylome to changes in gene expression and susceptibility-associated regions further indicates that hypomethylated regions are significantly associated with the up-regulation of cell activation transcriptional programs. Altogether, these findings implicate aberrant B cell function in MS etiology.


Asunto(s)
Linfocitos B/metabolismo , Activación de Linfocitos , Esclerosis Múltiple/metabolismo , Linfocitos B/patología , Diferenciación Celular , Metilación de ADN , Epigénesis Genética , Epigenómica , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Activación Transcripcional
4.
Immunol Cell Biol ; 101(4): 358-367, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36681886

RESUMEN

B cells play a key mechanistic role in the pathogenesis of multiple sclerosis (MS), a chronic neurological disease of the central nervous system with an autoimmune etiology. B cells contribute to disease initiation and progression by acting as professional antigen-presenting cells as well as via secreting autoantibodies and proinflammatory cytokines. We have recently shown that the polyglutamine protein ataxin-1, which was first linked to the movement disorder spinocerebellar ataxia type 1, also acts as a master regulator of B-cell functions in the context of central nervous system autoimmunity. In fact, ataxin-1-deficient mice display an aggravated manifestation of the MS disease model experimental autoimmune encephalomyelitis along with aberrant B-cell functions. Consistent with this scenario, transcriptomic analysis of Atxn1-null B cells highlighted distinct genetic signatures involved in cell activation, proliferation and antigen presentation. To further characterize the role of ataxin-1, we profiled the noncoding transcriptome controlled by ataxin-1 in the B-cell compartment upon an encephalitogenic challenge. We show that two specific classes of noncoding RNAs, namely, processed pseudogenes and intergenic long noncoding RNAs, are differentially regulated along disease. Furthermore, pathway and protein network analyses on their putative protein-coding gene targets found a significant enrichment in ontologies related to cell mitosis, together with molecular processes relevant to MS such as chitin metabolism. Altogether, these findings shed light on the possible contribution of noncoding RNAs to B-cell biology and MS pathogenesis, and further establish the immunomodulatory role of ataxin-1 in autoimmune demyelination.


Asunto(s)
Ataxina-1 , Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Ataxias Espinocerebelosas , Animales , Ratones , Ataxina-1/genética , Sistema Nervioso Central , Encefalomielitis Autoinmune Experimental/genética , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo
5.
Mol Cell Neurosci ; 120: 103707, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35231567

RESUMEN

The neuronal microtubule-associated protein tau undergoes multiple post-translational modifications, which dynamically modulate its molecular functions and biochemical features in space and time. Among them, we have recently reported that a conserved lysine residue mapping to the microtubule-binding domain of the protein (K306 in mouse and K317 in human) is differentially methylated in a model of chronic autoimmune demyelination. In contrast with other well-studied tau post-translational modifications such as phosphorylation, lysine methylation is far less investigated and its specific impact on tau biology is not fully understood. Here we performed a comprehensive analysis of the effects of K317 methylation on key tau features. By combining in silico simulations with in vitro biochemical assays and live-cell imaging, we show that methylated tau is more prone to self-assembly into insoluble structures. Moreover, we demonstrate that K317 methylation affects the stabilization activity of tau on microtubule dynamics. Lastly, we highlight a role for K317 methylation in regulating both neuronal differentiation and cell proliferation. Altogether, these findings shed light on the biology of an overlooked tau post-translational modification as well as on the fine tuning of tau functionality in health and disease.


Asunto(s)
Lisina , Proteínas tau , Animales , Lisina/metabolismo , Metilación , Ratones , Neuronas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas tau/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(38): 23742-23750, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32878998

RESUMEN

Ataxin-1 (ATXN1) is a ubiquitous polyglutamine protein expressed primarily in the nucleus where it binds chromatin and functions as a transcriptional repressor. Mutant forms of ataxin-1 containing expanded glutamine stretches cause the movement disorder spinocerebellar ataxia type 1 (SCA1) through a toxic gain-of-function mechanism in the cerebellum. Conversely, ATXN1 loss-of-function is implicated in cancer development and Alzheimer's disease (AD) pathogenesis. ATXN1 was recently nominated as a susceptibility locus for multiple sclerosis (MS). Here, we show that Atxn1-null mice develop a more severe experimental autoimmune encephalomyelitis (EAE) course compared to wildtype mice. The aggravated phenotype is mediated by increased T helper type 1 (Th1) cell polarization, which in turn results from the dysregulation of B cell activity. Ataxin-1 ablation in B cells leads to aberrant expression of key costimulatory molecules involved in proinflammatory T cell differentiation, including cluster of differentiation (CD)44 and CD80. In addition, comprehensive phosphoflow cytometry and transcriptional profiling link the exaggerated proliferation of ataxin-1 deficient B cells to the activation of extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription (STAT) pathways. Lastly, selective deletion of the physiological binding partner capicua (CIC) demonstrates the importance of ATXN1 native interactions for correct B cell functioning. Altogether, we report a immunomodulatory role for ataxin-1 and provide a functional description of the ATXN1 locus genetic association with MS risk.


Asunto(s)
Ataxina-1/metabolismo , Linfocitos B/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Animales , Presentación de Antígeno , Proliferación Celular , Encefalomielitis Autoinmune Experimental/fisiopatología , Ratones , Ratones Noqueados , Esclerosis Múltiple , Transducción de Señal
7.
Arch Biochem Biophys ; 719: 109156, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35218721

RESUMEN

The human leukocyte antigen (HLA) locus encodes a large group of proteins governing adaptive and innate immune responses. Among them, HLA class II proteins form α/ß heterodimers on the membrane of professional antigen-presenting cells (APCs), where they display both, self and pathogen-derived exogenous antigens to CD4+ T lymphocytes. We have previously shown that a shorter HLA-DRA isoform (sHLA-DRA) lacking 25 amino acids can be presented onto the cell membrane via binding to canonical HLA-DR2 heterodimers. Here, we employed atomistic molecular dynamics simulations to decipher the binding position of sHLA-DRA and its structural impact on functional regions of the HLA-DR2 molecule. We show that a loop region exposed only in the short isoform (residues R69 to G83) is responsible for binding to the outer domain of the HLA-DR2 peptide-binding site, and experimentally validated the critical role of F76 in mediating such interaction. Additionally, sHLA-DRA allosterically modifies the peptide-binding pocket conformation. In summary, this study unravels key molecular mechanisms underlying sHLA-DRA function, providing important insights into the role of full-length proteins in structural modulation of HLA class II receptors.


Asunto(s)
Antígeno HLA-DR2 , Péptidos , Sitios de Unión , Cadenas alfa de HLA-DR , Antígeno HLA-DR2/química , Antígeno HLA-DR2/metabolismo , Humanos , Isoformas de Proteínas/metabolismo
8.
Immunology ; 162(2): 194-207, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32986852

RESUMEN

Class II human leucocyte antigen (HLA) proteins are involved in the immune response by presenting pathogen-derived peptides to CD4+ T lymphocytes. At the molecular level, they are constituted by α/ß-heterodimers on the surface of professional antigen-presenting cells. Here, we report that the acceptor variant (rs8084) in the HLA-DRA gene mediates the transcription of an alternative version of the α-chain lacking 25 amino acids in its extracellular domain. Molecular dynamics simulations suggest this isoform undergoes structural refolding which in turn affects its stability and cellular trafficking. The short HLA-DRA isoform cannot reach the cell surface, although it is still able to bind the corresponding ß-chain. Conversely, it remains entrapped within the endoplasmic reticulum where it is targeted for degradation. Furthermore, we demonstrate that the short isoform can be transported to the cell membrane via interactions with the peptide-binding site of canonical HLA heterodimers. Altogether, our findings indicate that short HLA-DRA functions as a novel intact antigen for class II HLA molecules.


Asunto(s)
Cadenas alfa de HLA-DR/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Isoformas de Proteínas/inmunología , Adulto , Anciano , Aminoácidos/inmunología , Células Presentadoras de Antígenos/inmunología , Sitios de Unión/inmunología , Línea Celular , Línea Celular Tumoral , Membrana Celular/inmunología , Retículo Endoplásmico/inmunología , Femenino , Células HEK293 , Células HeLa , Humanos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Péptidos/inmunología , Linfocitopenia-T Idiopática CD4-Positiva/inmunología
9.
Hum Mol Genet ; 28(8): 1343-1356, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30590536

RESUMEN

Dystonia is a movement disorder characterized by involuntary and repetitive co-contractions of agonist and antagonist muscles. Dystonia 6 (DYT6) is an autosomal dominant dystonia caused by loss-of-function mutations in the zinc finger transcription factor THAP1. We have generated Thap1 knock-out mice with a view to understanding its transcriptional role. While germ-line deletion of Thap1 is embryonic lethal, mice lacking one Thap1 allele-which in principle should recapitulate the haploinsufficiency of the human syndrome-do not show a discernable phenotype. This is because mice show autoregulation of Thap1 mRNA levels with upregulation at the non-affected locus. We then deleted Thap1 in glial and neuronal precursors using a nestin-conditional approach. Although these mice do not exhibit dystonia, they show pronounced locomotor deficits reflecting derangements in the cerebellar and basal ganglia circuitry. These behavioral features are associated with alterations in the expression of genes involved in nervous system development, synaptic transmission, cytoskeleton, gliosis and dopamine signaling that link DYT6 to other primary and secondary dystonic syndromes.


Asunto(s)
Proteínas de Unión al ADN/genética , Distonía Muscular Deformante/genética , Trastornos Distónicos/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al ADN/fisiología , Modelos Animales de Enfermedad , Distonía/genética , Distonía Muscular Deformante/fisiopatología , Trastornos Distónicos/fisiopatología , Regulación de la Expresión Génica/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Síndrome , Dedos de Zinc
10.
Genes Immun ; 21(5): 288-300, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33011744

RESUMEN

Tau is an evolutionary conserved protein that promotes the assembly and stabilization of microtubules in neuronal axons. Complex patterns of posttranslational modifications (PTMs) dynamically regulate tau biochemical properties and consequently its functions. An imbalance in tau PTMs has been connected with a broad spectrum of neurodegenerative conditions which are collectively known as tauopathies and include Alzheimer's disease (AD), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD) among others. The hallmark of these neurological disorders is the presence in the brain of fibrillary tangles constituted of misfolded species of hyper-phosphorylated tau. The pathological events leading to tau aggregation are still largely unknown but increasing evidence suggests that neuroinflammation plays a critical role in tangle formation. Moreover, tau aggregation itself could enhance inflammation through feed-forward mechanisms, amplifying the initial neurotoxic insults. Protective effects of tau against neuroinflammation have been also documented, adding another layer of complexity to this phenomenon. Here, we will review the current knowledge on tau regulation and function in health and disease. In particular, we will address its emerging role in connecting neurodegenerative and neuroinflammatory processes.


Asunto(s)
Tauopatías/inmunología , Proteínas tau/metabolismo , Animales , Humanos , Neuroglía/inmunología , Linfocitos T/inmunología , Tauopatías/genética , Tauopatías/patología , Proteínas tau/genética
11.
J Neuroinflammation ; 17(1): 297, 2020 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046105

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) belong to a class of evolutionary conserved, non-coding small RNAs with regulatory functions on gene expression. They negatively affect the expression of target genes by promoting either RNA degradation or translational inhibition. In recent years, converging studies have identified miRNAs as key regulators of oligodendrocyte (OL) functions. OLs are the cells responsible for the formation and maintenance of myelin in the central nervous system (CNS) and represent a principal target of the autoimmune injury in multiple sclerosis (MS). METHODS: MiRAP is a novel cell-specific miRNA affinity-purification technique which relies on genetically tagging Argonaut 2 (AGO2), an enzyme involved in miRNA processing. Here, we exploited miRAP potentiality to characterize OL-specific miRNA dynamics in the MS model experimental autoimmune encephalomyelitis (EAE). RESULTS: We show that 20 miRNAs are differentially regulated in OLs upon transition from pre-symptomatic EAE stages to disease peak. Subsequent in vitro differentiation experiments demonstrated that a sub-group of them affects the OL maturation process, mediating either protective or detrimental signals. Lastly, transcriptome profiling highlighted the endocytosis, ferroptosis, and FoxO cascades as the pathways associated with miRNAs mediating or inhibiting OL maturation. CONCLUSIONS: Altogether, our work supports a dual role for miRNAs in autoimmune demyelination. In particular, the enrichment in miRNAs mediating pro-myelinating signals suggests an active involvement of these non-coding RNAs in the homeostatic response toward neuroinflammatory injury.


Asunto(s)
Proteínas Argonautas/biosíntesis , Encefalomielitis Autoinmune Experimental/metabolismo , Perfilación de la Expresión Génica/métodos , MicroARNs/biosíntesis , Oligodendroglía/metabolismo , Animales , Proteínas Argonautas/genética , Células Cultivadas , Encefalomielitis Autoinmune Experimental/genética , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética
12.
Proc Natl Acad Sci U S A ; 114(33): E6982-E6991, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28760957

RESUMEN

Proinflammatory mononuclear phagocytes (MPs) play a crucial role in the progression of multiple sclerosis (MS) and other neurodegenerative diseases. Despite advances in neuroimaging, there are currently limited available methods enabling noninvasive detection of MPs in vivo. Interestingly, upon activation and subsequent differentiation toward a proinflammatory phenotype MPs undergo metabolic reprogramming that results in increased glycolysis and production of lactate. Hyperpolarized (HP) 13C magnetic resonance spectroscopic imaging (MRSI) is a clinically translatable imaging method that allows noninvasive monitoring of metabolic pathways in real time. This method has proven highly useful to monitor the Warburg effect in cancer, through MR detection of increased HP [1-13C]pyruvate-to-lactate conversion. However, to date, this method has never been applied to the study of neuroinflammation. Here, we questioned the potential of 13C MRSI of HP [1-13C]pyruvate to monitor the presence of neuroinflammatory lesions in vivo in the cuprizone mouse model of MS. First, we demonstrated that 13C MRSI could detect a significant increase in HP [1-13C]pyruvate-to-lactate conversion, which was associated with a high density of proinflammatory MPs. We further demonstrated that the increase in HP [1-13C]lactate was likely mediated by pyruvate dehydrogenase kinase 1 up-regulation in activated MPs, resulting in regional pyruvate dehydrogenase inhibition. Altogether, our results demonstrate a potential for 13C MRSI of HP [1-13C]pyruvate as a neuroimaging method for assessment of inflammatory lesions. This approach could prove useful not only in MS but also in other neurological diseases presenting inflammatory components.


Asunto(s)
Isótopos de Carbono , Ácido Láctico , Imagen por Resonancia Magnética , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/metabolismo , Animales , Isótopos de Carbono/farmacocinética , Isótopos de Carbono/farmacología , Cuprizona/efectos adversos , Cuprizona/farmacología , Modelos Animales de Enfermedad , Femenino , Ácido Láctico/farmacocinética , Ácido Láctico/farmacología , Ratones , Ratones Transgénicos , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/genética
13.
J Autoimmun ; 101: 56-69, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31010726

RESUMEN

The molecular events underlying the transition from initial inflammatory flares to the progressive phase of multiple sclerosis (MS) remain poorly understood. Here, we report that the microtubule-associated protein (MAP) Tau exerts a gender-specific protective function on disease progression in the MS model experimental autoimmune encephalomyelitis (EAE). A detailed investigation of the autoimmune response in Tau-deficient mice excluded a strong immunoregulatory role for Tau, suggesting that its beneficial effects are presumably exerted within the central nervous system (CNS). Spinal cord transcriptomic data show increased synaptic dysfunctions and alterations in the NF-kB activation pathway upon EAE in Tau-deficient mice as compared to wildtype animals. We also performed the first comprehensive characterization of Tau post-translational modifications (PTMs) in the nervous system upon EAE. We report that the methylation levels of the conserved lysine residue K306 are significantly decreased in the chronic phase of the disease. By combining biochemical assays and molecular dynamics (MD) simulations, we demonstrate that methylation at K306 decreases the affinity of Tau for the microtubule network. Thus, the down-regulation of this PTM might represent a homeostatic response to enhance axonal stability against an autoimmune CNS insult. The results, altogether, position Tau as key mediator between the inflammatory processes and neurodegeneration that seems to unify many CNS diseases.


Asunto(s)
Regulación de la Expresión Génica , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Neuronas/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Proteínas tau/metabolismo , Animales , Autoinmunidad , Línea Celular , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental , Femenino , Redes Reguladoras de Genes , Masculino , Metilación , Ratones , Ratones Noqueados , Modelos Moleculares , Esclerosis Múltiple/patología , Transducción de Señal , Relación Estructura-Actividad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Transcripción Genética , Proteínas tau/química
14.
J Neuroinflammation ; 15(1): 72, 2018 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-29514694

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is characterized by increased activation of peripheral blood mononuclear cells (PBMCs), linked to perturbations in the phosphorylation of signaling proteins. METHODS: We developed a phosphoflow cytometry protocol to assess the levels of 11 phosphorylated nuclear proteins at baseline conditions and after cell activation in distinct PBMC populations from 41 treatment-naïve relapsing-remitting (RR) MS subjects and 37 healthy controls, and in a second cohort of 9 untreated RRMS patients and 10 secondary progressive (SP) MS patients. Levels of HLA-ABC, HLA-E, and HLA-DR were also assessed. Phosphorylation levels of selected proteins were also assessed in mouse splenocytes isolated from myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis (EAE). RESULTS: Modest differences were observed at baseline between patients and controls, with general lower phosphorylation levels in cells from affected individuals. Conversely, a dramatic increase in phosphorylated p38MAPK and STAT proteins was observed across all cell types in MS patients compared to controls after in vitro activation. A similar phosphorylation profile was observed in mouse lymphocytes primed in vivo with MOG. Furthermore, levels of all p-STAT proteins were found directly correlated with HLA expression in monocytes. Levels of phosphorylated proteins did not differ between relapsing-remitting and secondary progressive MS patients either in baseline conditions or after stimulation. Lastly, phosphorylation levels appear to be independent of the genotype. CONCLUSION: The response to IFN-α through STAT proteins signaling is strongly dysregulated in MS patients irrespective of disease stage. These findings suggest that the aberrant activation of this pathway could lead to changes in the expression of HLA molecules in antigen presenting cells, which are known to play important roles in the regulation of the immune response in health and disease.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Esclerosis Múltiple/patología , Factores de Transcripción STAT/metabolismo , Transducción de Señal/fisiología , Adulto , Animales , Estudios de Casos y Controles , Estudios de Cohortes , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Femenino , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fosforilación
15.
Hum Mol Genet ; 24(24): 7151-8, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26433934

RESUMEN

Despite recent progress in the characterization of genetic loci associated with multiple sclerosis (MS) risk, the ubiquitous linkage disequilibrium operating across the genome has stalled efforts to distinguish causative variants from proxy single-nucleotide polymorphisms (SNPs). Here, we have identified through fine mapping and meta-analysis EVI5 as the most plausible disease risk gene within the 1p22.1 locus. We further show that an exonic SNP associated with risk induces changes in superficial hydrophobicity patterns of the coiled-coil domain of EVI5, which, in turns, affects the EVI5 interactome. Immunoprecipitation of wild-type and mutated EVI5 followed by mass spectrometry generated a roster of disease-specific interactors functionally linked to lipid metabolism. Among the exclusive binding partners of the risk variant, we describe the novel interaction with sphingosine 1-phosphate lyase (SGPL1)-a key enzyme for the creation of the sphingosine-1 phosphate gradient, which is relevant to the pathogenic process and therapeutic management of MS.


Asunto(s)
Esclerosis Múltiple/genética , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple , Proteínas de Ciclo Celular , Mapeo Cromosómico , Cromosomas Humanos Par 1 , Proteínas Activadoras de GTPasa , Células HeLa , Humanos , Proteoma/metabolismo , Factores de Riesgo
16.
Hum Mol Genet ; 23(14): 3733-45, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24594842

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is an incurable neurodegenerative disease caused by a pathogenic glutamine repeat expansion in the protein ataxin-1 (ATXN1). One likely mechanism mediating pathogenesis is excessive transcriptional repression induced by the expanded ATXN-1. Because ATXN1 binds HDAC3, a Class I histone deacetylase (HDAC) that we have found to be required for ATXN1-induced transcriptional repression, we tested whether genetically depleting HDAC3 improves the phenotype of the SCA1 knock-in mouse (SCA1(154Q/2Q)), the most physiologically relevant model of SCA1. Given that HDAC3 null mice are embryonic lethal, we used for our analyses a combination of HDAC3 haploinsufficient and Purkinje cell (PC)-specific HDAC3 null mice. Although deleting a single allele of HDAC3 in the context of SCA1 was insufficient to improve cerebellar and cognitive deficits of the disease, a complete loss of PC HDAC3 was highly deleterious both behaviorally, with mice showing early onset ataxia, and pathologically, with progressive histologic evidence of degeneration. Inhibition of HDAC3 may yet have a role in SCA1 therapy, but our study provides cautionary evidence that this approach could produce untoward effects. Indeed, the neurotoxic consequences of HDAC3 depletion could prove relevant, wherever pharmacologic inhibition of HDAC3 is being contemplated, in disorders ranging from cancer to neurodegeneration.


Asunto(s)
Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Células de Purkinje/metabolismo , Ataxias Espinocerebelosas/patología , Animales , Ataxina-1 , Ataxinas , Peso Corporal , Línea Celular Tumoral , Cerebelo/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Células HEK293 , Haploinsuficiencia , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Actividad Motora , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Células de Purkinje/patología , Ataxias Espinocerebelosas/tratamiento farmacológico , Ataxias Espinocerebelosas/genética
17.
J Neuroimmunol ; 391: 578366, 2024 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-38733741

RESUMEN

Disturbance in neuroendocrine signaling has been consistently documented in multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS) representing the main cause of non-traumatic brain injury among young adults. In fact, MS patients display altered hormonal levels and psychiatric symptoms along with the pathologic hallmarks of the disease, which include demyelination, neuroinflammation and axonal injury. In addition, we have recently shown that extensive transcriptional changes take place in the hypothalamus of mice upon the MS model experimental autoimmune encephalomyelitis (EAE). We also detected structural and functional aberrancies in endocrine glands of EAE animals. Specifically, we described the hyperplasia of adrenal glands and the atrophy of ovaries at disease peak. To further expand the characterization of these phenotypes, here we profiled the transcriptomes of both glands by means of RNA-seq technology. Notably, we identified fatty acid and cholesterol biosynthetic pathways as the most dysregulated molecular processes in adrenals and ovaries, respectively. Furthermore, we demonstrated that key genes encoding neuropeptides and hormone receptors undergo distinct expression dynamics in the hypothalamus along disease progression. Altogether, our results corroborate the dysfunction of the neuroendocrine system as a major pathological event of autoimmune demyelination and highlight the crosstalk between the CNS and the periphery in mediating such disease phenotypes.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Metabolismo de los Lípidos , Ratones Endogámicos C57BL , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Metabolismo de los Lípidos/fisiología , Ovario/metabolismo , Ovario/patología , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/patología , Transcriptoma
18.
Cell Mol Biol Lett ; 18(2): 209-30, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23479001

RESUMEN

Prion diseases are a class of fatal neurodegenerative disorders that can be sporadic, genetic or iatrogenic. They are characterized by the unique nature of their etiologic agent: prions (PrP(Sc)). A prion is an infectious protein with the ability to convert the host-encoded cellular prion protein (PrP(C)) into new prion molecules by acting as a template. Since Stanley B. Prusiner proposed the "protein-only" hypothesis for the first time, considerable effort has been put into defining the role played by PrP(C) in neurons. However, its physiological function remains unclear. This review summarizes the major findings that support the involvement of PrP(C) in signal transduction.


Asunto(s)
Priones/metabolismo , Transducción de Señal , Animales , Técnicas de Inactivación de Genes , Humanos , Ligandos , Modelos Biológicos , Priones/química , Proteínas Quinasas/metabolismo
19.
J Vis Exp ; (202)2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38108416

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is a disease model that recapitulates the autoimmune disorder multiple sclerosis (MS) at histopathological and molecular levels. EAE is induced by immunizing experimental animals via subcutaneous injection of short myelin peptides together with specific adjuvants to boost the immune response. Like the human counterpart, EAE mice develop demyelinating lesions, immune cell infiltration into the central nervous system (CNS), glia activation and neuronal injury. A consistent body of evidence also supports a mechanistic role for B cell dysfunction in the etiology of both MS and EAE. B cells can serve as antigen-presenting cells as well as a primary source of pro-inflammatory cytokines and autoantibodies. In EAE, antibodies are generated against the myelin peptides that were employed to induce the disease. Such autoantibodies have been shown to mediate either myelin loss or pathogenic T cell reactivation into the CNS. This article describes an efficient ELISA-based protocol to quantify autoantibodies in the serum of C57BL/6J mice immunized with the myelin oligodendrocyte glycoprotein 35-55 (MOG35-55) peptide. The proposed method serves as a powerful tool to investigate the specificity and magnitude of the aberrant humoral response in the context of autoimmune demyelination.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Humanos , Animales , Ratones , Ratones Endogámicos C57BL , Autoanticuerpos , Sistema Nervioso Central , Citocinas
20.
Commun Biol ; 6(1): 342, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36997638

RESUMEN

Genome-wide association studies (GWAS) successfully identified multiple sclerosis (MS) susceptibility variants. Despite this notable progress, understanding the biological context of these associations remains challenging, due in part to the complexity of linking GWAS results to causative genes and cell types. Here, we aimed to address this gap by integrating GWAS data with single-cell and bulk chromatin accessibility data and histone modification profiles from immune and nervous systems. MS-GWAS associations are significantly enriched in regulatory regions of microglia and peripheral immune cell subtypes, especially B cells and monocytes. Cell-specific polygenic risk scores were developed to examine the cumulative impact of the susceptibility genes on MS risk and clinical phenotypes, showing significant associations with risk and brain white matter volume. The findings reveal enrichment of GWAS signals in B cell and monocyte/microglial cell-types, consistent with the known pathology and presumed targets of effective MS therapeutics.


Asunto(s)
Linfocitos B , Microglía , Monocitos , Esclerosis Múltiple , Humanos , Linfocitos B/metabolismo , Células Sanguíneas/metabolismo , Cromatina , Elementos de Facilitación Genéticos , Epigénesis Genética , Predisposición Genética a la Enfermedad , Puntuación de Riesgo Genético , Variación Genética , Microglía/metabolismo , Monocitos/metabolismo , Esclerosis Múltiple/genética , Análisis de Expresión Génica de una Sola Célula , Encéfalo/citología , Biobanco del Reino Unido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA